Next Article in Journal
Deciphering the Potentials of Cardamom in Cancer Prevention and Therapy: From Kitchen to Clinic
Next Article in Special Issue
The Imbalance of Homocysteine, Vitamin B12 and Folic Acid in Parkinson Plus Syndromes: A Review beyond Parkinson Disease
Previous Article in Journal
Are Small Molecules Effective in Treating Inflammatory Pouch Disorders Following Ileal Pouch-Anal Anastomosis for Ulcerative Colitis? Here Is Where We Stand
Previous Article in Special Issue
How the Western Diet Thwarts the Epigenetic Efforts of Gut Microbes in Ulcerative Colitis and Its Association with Colorectal Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Complex Pathophysiology of Acute Kidney Injury (AKI) in Aging: Epigenetic Regulation, Matrix Remodeling, and the Healing Effects of H2S

by
Shreyasi Gupta
1,
Subhadeep Mandal
1,
Kalyan Banerjee
1,
Hebah Almarshood
2,
Sathnur B. Pushpakumar
2 and
Utpal Sen
2,*
1
Department of Zoology, Trivenidevi Bhalotia College, College Para Rd, Raniganj 713347, West Bengal, India
2
Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
*
Author to whom correspondence should be addressed.
Biomolecules 2024, 14(9), 1165; https://doi.org/10.3390/biom14091165
Submission received: 12 May 2024 / Revised: 12 September 2024 / Accepted: 14 September 2024 / Published: 17 September 2024
(This article belongs to the Special Issue Homocysteine and H2S in Health and Disease)

Abstract

:
The kidney is an essential excretory organ that works as a filter of toxins and metabolic by-products of the human body and maintains osmotic pressure throughout life. The kidney undergoes several physiological, morphological, and structural changes with age. As life expectancy in humans increases, cell senescence in renal aging is a growing challenge. Identifying age-related kidney disorders and their cause is one of the contemporary public health challenges. While the structural abnormalities to the extracellular matrix (ECM) occur, in part, due to changes in MMPs, EMMPRIN, and Meprin-A, a variety of epigenetic modifiers, such as DNA methylation, histone alterations, changes in small non-coding RNA, and microRNA (miRNA) expressions are proven to play pivotal roles in renal pathology. An aged kidney is vulnerable to acute injury due to ischemia-reperfusion, toxic medications, altered matrix proteins, systemic hemodynamics, etc., non-coding RNA and miRNAs play an important role in renal homeostasis, and alterations of their expressions can be considered as a good marker for AKI. Other epigenetic changes, such as histone modifications and DNA methylation, are also evident in AKI pathophysiology. The endogenous production of gaseous molecule hydrogen sulfide (H2S) was documented in the early 1980s, but its ameliorative effects, especially on kidney injury, still need further research to understand its molecular mode of action in detail. H2S donors heal fibrotic kidney tissues, attenuate oxidative stress, apoptosis, inflammation, and GFR, and also modulate the renin–angiotensin–aldosterone system (RAAS). In this review, we discuss the complex pathophysiological interplay in AKI and its available treatments along with future perspectives. The basic role of H2S in the kidney has been summarized, and recent references and knowledge gaps are also addressed. Finally, the healing effects of H2S in AKI are described with special emphasis on epigenetic regulation and matrix remodeling.

1. Introduction

Acute kidney injury (AKI) is a pathophysiological condition that can be diagnosed by elevated creatinine levels and/or decreased urine output [1]. In developed countries, the incidence of severe AKI is almost 3.2 to 9.6% of total hospitalized patients, whereas in developing countries, AKI mortality rates are still as high as 50–60% [2] and need much more attention. AKI causes approximately 2 million fatalities annually worldwide [3,4] and affects up to 50% of critically ill intensive care unit (ICU) patients as their severity increases mortality rates [5,6]. Patients who are old or have a prior disease history, such as chronic kidney disease (CKD), diabetes, cancer, etc., are more vulnerable to form AKI. However, several molecular mechanisms underlying different stages of AKI pathogenesis are still a matter of conjecture and very patient-specific. It is therefore important to identify novel treatment targets and early diagnostic indicators to reduce the mortality rate. One of the major causes of AKI is ischemia-reperfusion (I/R) damage [7,8]. I/R causes inflammatory cells like macrophages to infiltrate within kidney tissue from blood vessels and induce an immunological inflammatory response that leads to the onset of AKI [9]. Moreover, patients who have survived an AKI are more likely to develop CKD [10,11,12].
Age plays a crucial role in kidney health. The replenishment of renal parenchyma significantly decreases in geriatric conditions, and fibrosis takes place due to excessive extracellular matrix (ECM) production in the kidney interstitium [13]. Declined renal function with age is multidimensional and undoubtedly leads to susceptibility to AKI [14], especially nephrotoxic AKI and I/R AKI, and also a declined GFR and healthy nephron number, and tubular epithelial cells [15,16,17]. Cohort studies showed that dialysis is required in AKI in elderly patients with a significantly higher percentage [18,19].
Hydrogen sulfide (H2S) is one of the endogenous gasotransmitters and is involved in a variety of biological processes [20,21]. The study suggests that H2S donors can regulate the rate of blood pressure, oxidative damage, inducible NO synthase, NF-kβ activation, and apoptosis in damaged and aged kidneys [22,23,24,25,26,27,28,29,30,31,32] (Figure 1). The possible healing functions of H2S in AKI, e.g., regulating renin release, oxygen sensing, salt absorption, and the glomerular filtration rate (GFR), in association with aging, have been evident in several research [33,34,35,36,37,38,39,40]. H2S donors are reported to ameliorate aging-induced kidney changes probably by inhibiting the IR/IRS-2-Akt-mTORC1 signaling pathways, leading to matrix protein synthesis [41].
In this review, we discuss the recent advances in experimental as well as preclinical and clinical research in renal aging and AKI with special references to different epigenetic regulations, such as DNA methylation, histone alterations, non-coding RNA regulation, and/or changes in microRNA. We also discuss how the H2S level significantly facilitates the production of the renal matrix in association with EMMPRIN, Meprin-A, and MMPs and helps healing. In the final part, current treatment modalities and therapeutic options with future possibilities and challenges are summarized.

2. Production and Detection of H2S

Hydrogen sulfide (H2S) altered N-methyl-D-aspartate (NMDA) receptor-mediated functions in the hippocampus, and thus, the physiological significance of H2S first became apparent in 1996 [43]. Endogenous H2S serves several pleiotropic functions within the cardiovascular system, kidneys, liver, pancreas, gastrointestinal system, brain, and reproductive system, including others [34,44,45,46,47,48]. The enzymes responsible for H2S synthesis are the following: cystathionine β-synthase (CBS) [49], cystathionine γ-lyase (CSE) [50,51], and 3-mercaptopyruvate sulfurtransferase (3-MST) [52]. H2S is present in blood from the nanomolar to micromolar range and thus can travel to different tissues as per requirement [53], although it was further assumed that the micromolar range is a bit of an overestimation due to the limitations in the detection methods [54]. However, regardless of its concentration in the blood, plasma, or tissue, it is a well-recognized gaseous transmitter that has immense physiological effects [54].
Kidneys are a major producer of H2S [38,55], in addition to the liver, brain, heart, skeletal muscle, and other major tissues [39]. Under physiological conditions, all three enzymes, i.e., CBS, CSE, and 3-MST, are involved in H2S production, with CBS and CSE being the major contributors in the kidney [38,39]. It is reported that 3-MST acts in conjunction with cysteine aminotransferase [56]. In the mitochondria, H2S is produced by the reduction of thiosulfate catalyzed by thiosulfate transferase (TST) [56]. Biochemical studies have shown that kidneys are a rich source of enzymatic generation of H2S by all five enzymes [56,57].
H2S is produced enzymatically by two different mechanisms, including pyridoxal 5′-phosphate-dependent (CBS and CSE) and pyridoxal 5′-phosphate-independent (3-MST) systems [21,58,59]. In renal tissue, it has been noted that CBS primarily localizes to the proximal convoluted tubule in the outer cortex [60,61]. According to the reports, the CSE enzyme is localized in the glomerulus and tubulointerstitial region [62]. In addition, 3-MST is localized in the proximal tubular epithelium [63]. CBS mediates the conversion of l-homocysteine (Hcy) and L-Serine to produce cystathionine, which is then converted by CSE into l-cysteine [64,65]. Furthermore, Hcy is converted to homolanthionine by CSE, which catalyzes the formation of H2S [66]. Finally, aspartate aminotransferase uses α-ketoglutarate to convert cysteine into 3-mercaptopyruvate and glutamate. In the presence of a reductant, 3-mercaptopyruvate sulfur transferase (3-MST), an additional enzyme, uses 3-mercaptopyruvate as its substrate to contribute to the endogenous synthesis of H2S in the mitochondria [52,67]. Further research revealed that 3-MST and D-amino acid oxidase (DAO) are involved in the production of H2S from D-cysteine [68]. D-serine is thought to create more H2S in the kidney than L-serine [68]. A schematic diagram is shown in Figure 2.
The detection of H2S levels in blood or tissue requires an accurate, sensitive, and analytical method [69,70]. Researchers have developed methods for detecting H2S, such as gas chromatography, electrochemistry, high-performance liquid chromatography, inductively coupled plasma-atomic emission, UV–visible absorption spectrometry, and fluorescence spectrometry [69,71,72,73]. Compared to other detection methods, electrochemical sensors offer high sensitivity and convenience in H2S detection, while fluorescent and colorimetric sensors provide a fast response time, high efficiency and low cost, visualization, and better in vivo imaging [69,72,74,75,76,77,78]. Combining these advantages could improve H2S sensor performance and open up new ways to further explore novel H2S chemical sensors with excellent properties. Advanced chemical sensors for a highly sensitive real-time visual detection are a possibility as a result of future developments in nanomaterials, fluorescent dyes, and high-precision detection technologies [69].

3. The Function of H2S in Kidney

Hydrogen sulfide (H2S) plays a crucial role in maintaining physiological homeostasis through various mechanisms [79]. It regulates kinases, ion channels, and transcription factors by S-sulfhydrating cysteine residues. Additionally, H2S binds to heme in heme-containing proteins, acts as a free radical scavenger, and donates electrons to the mitochondrial electron transport chain, enhancing mitochondrial ATP production and regulating bioenergetics [36,45,80,81,82]. The functions of H2S are tissue-specific and we focused on summarizing its role in the kidney below.

3.1. H2S Effect on Renal Excretory Function and Water Handling

It has been demonstrated that H2S alters cellular function in the kidney in a number of ways, with a range of downstream implications. The intrarenal injection of donor H2S, NaHS can raise the glomerular filtration rate (GFR), urinary sodium (Una.V), and potassium excretion (Uk.V) [34,35]. Moreover, the effect is closely mimicked by the infusion of L-cysteine, an H2S-generating substrate [34].
In addition, the inhibition of endogenous H2S production by aminooxyacetic acid (AOAA) (CBS inhibitor) plus DL-propargylglycine (PAG) (CSE inhibitor) causes GFR, Una.V, and Uk.V to decline. This finding indicates that under physiological conditions, H2S controls renal function [39]. Another investigation has established that there is a compensating effect between CBS and CSE on renal control, while neither AOAA nor PAG alone can have any effect on renal function [39,83].
In summary, H2S promotes vasodilation and inhibits the activities of Na+/K+-ATPase (NKA) and the Na+-K+-Cl-cotransporter (NKCC), which in turn raises renal blood flow (RBF) and GFR and encourages natriuresis (UNa.V) and kaliuresis (Uk.V) [36] (Figure 3A). However, it is still unclear how H2S affects the kidney’s ionic exchanger ability.
Several evidence suggest that H2S is involved in renal water handling [36,84]. Renal aquaporin 2 (AQP2) expression is upregulated when H2S activates the intracellular cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) signaling pathway [36,85]. This improves urine concentration and lowers urine osmolality in conditions affecting water balance, such as nephrogenic diabetic insipidus [36,84,85].

3.2. H2S Acts as an O2 Sensor in the Kidney

H2S serves as an oxygen sensor in the renal medulla during hypoxia, whereupon its production rises, promoting oxygen restoration, raising RBF and GFR, and inhibiting tubular transit [36]. While H2S is produced without oxygen, its oxidative metabolism in mitochondria requires oxygen [86]. The availability of O2 in renal medulla is lower compared with that in the renal cortex, resulting in a higher abundance of H2S in this region [87]. Therefore, the hypoxic environment caused by the low oxygen partial pressure in the renal medulla causes H2S to accumulate. This, in turn, increases H2S activity, including electron donation for ATP production in the mitochondria, and restores the oxygen balance by increasing the medullary flow and decreasing the tubular Na+ transport, which accounts for 60% of renal oxygen consumption [36,83,86,87,88,89,90,91] (Figure 3B). Furthermore, in hypoxic conditions, CBS and CSE can go into mitochondria and induce the synthesis of H2S [36,82,92]. However, more research is needed to fully understand the precise processes and subsequent signaling events.

3.3. H2S Modulates the Renin–Angiotensin–Aldosterone System (RAAS)

RAS activity is determined by the release of renin from juxtaglomerular cells (JG), and intracellular cAMP is known to modulate this process [39,93,94]. The renin–angioten–in–aldosterone system (RAAS) is a complex endocrine system that regulates electrolyte levels and blood volume to maintain blood pressure [95,96,97]. H2S has been shown to lower cAMP levels by inhibiting adenylyl cyclase (AC) in a variety of cell types, suggesting that it may control the release of renin, as well as reduce angiotensin II levels [39,98]. According to Lu, Ming et al., the H2S donor NaHS prevented and cured hypertension in the two-kidney and one-clip (2K1C) model of renovascular hypertension [98]. On the other hand, in rats that were normal or had one kidney, one clip (1K1C), both of which had normal plasma renin activity, NaHS had no effect on blood pressure or plasma renin activity [36,98]. In other studies, using primary cultures of renin-rich kidney cells, it was shown that treatment with 100 μmol/L of NaHS likewise markedly reduced the level of intracellular cAMP and repressed renin activity [36,98]. Laggner et al. showed that H2S directly reduces the activity of the angiotensin-converting enzyme (ACE) in a dose-dependent way [99]. In addition to AC, ROS was recently shown to be a target of H2S due to its impact on lowering renin levels in diabetic nephropathy (DN), indicating the involvement of several mechanisms in the process [36,39].

3.4. Aging Affects H2S Production

Our body naturally produces H2S, a gas that plays several roles in physiology, including controlling blood pressure, inflammation, the immune system, energy metabolism, and vascular function, among others [23,100]. Studies have also demonstrated the anti-aging [101] properties of H2S by demonstrating its antioxidant, anti-inflammatory [102,103], and cytoprotective [104] effects. One of the main contributing factors to aging and the emergence of age-related diseases is thought to be tissue damage by free radicals. In order to combat oxidative stress and possibly slow down the aging process, H2S has been shown to scavenge reactive oxygen species (ROS) [105] and increase the activity of antioxidant enzymes [106]. Pro-inflammatory cytokines, which are proteins that stimulate inflammation, are inhibited by H2S, which can thus reduce inflammation [104].
As we age, mitochondrial function declines, which can lead to a number of age-related problems. H2S can help to improve mitochondrial function by increasing the production of ATP [107,108]. The effects of aging may differ among tissues. It is known that the cardiovascular system is more susceptible to age-related changes than the liver [109,110]. Although the kidney undergoes structural and functional changes with aging, there are little data regarding the underlying mechanism [111]. However, it is not a surprise that due to age-related changes to the kidney, H2S production and signaling may also decline. A reduction in endogenous H2S production is reported to accelerate atherosclerosis, an age-dependent disease [112]. In fact, it has been demonstrated that inhibiting CBS expression in human umbilical vein endothelial cells causes premature aging [113]. Further, aging may reduce H2S production by causing oxidative damage to CBS/CSE proteins [114]. The above studies provide indirect evidence that a lack of H2S is associated with the aging process.

4. Epigenetic Regulation of Renal Aging and AKI

AKI may have multiple underlying causes and is linked to intricate pathophysiological processes [115,116,117]. In the last few decades, there has been a rise in the incidence of AKI [115,117]. Mortality rates have declined in critically ill patients with AKI, but mortality rates are still significantly high and increase with AKI severity, specifically in dialysis-requiring AKI [117,118,119,120,121].
The study of heritable mechanisms that regulate gene expression without altering the fundamental nucleotide sequence is known as epigenetics [122,123]. A variety of epigenetic modifiers, such as DNA methylation, histone alterations, non-coding RNA regulation, and/or changes in miRNA expressions are proven to have pivotal roles in molecular alterations during renal aging [124].

4.1. DNA Methylation and AKI

DNA methylation happens at the cytosines in CpG dinucleotides to generate 5-methylcytosine (5-mC) [125]; however, it can also occur at a low frequency in non-CpG sites, particularly in embryonic stem cells [126,127], oocytes [128], and brain tissues [129,130]. Sixty to ninety percent of CpG sites in the mammalian genome are methylated [124]. In eukaryotes, DNA methylation occurs only at cytosine residues and involves the covalent addition of a methyl group (CH3), which is commonly donated by S-adenosyl-L-methionine (SAM), to the 5-carbon position of the cytosine, creating 5-methylcytosine (5mC) via DNA methyltransferases (DNMTs) [131,132]. Three DNA methyltransferases (DNMTs)—DNMT1, DNMT3a, and DNMT3b—regularly create and maintain DNA methylation patterns in mammals [133,134,135].
In a recent study, Gao et al. found that administering DNA-demethylating agents, SGI-1027 and OLP, effectively alleviated D-gal-induced aging-related structural and functional alteration in mouse kidneys by reducing the DNA methylation of the anti-aging nuclear factor erythroid-derived 2-like 2 (NRF2) and KLOTHO promoter [135,136,137]. According to this study, certain gene expression patterns and genomic DNA methylation may actually have an impact on the aging process of the kidneys. In particular, the kidney aging process is greatly influenced by the dysregulation of DNMT1/3a/3b, and renal aging changes can be lessened by epigenetic intervention using DNA-demethylating agents [133,134,135,136,137]. The current information on DNA methylation changes in age-related kidney diseases is insufficient, necessitating future systematic studies and clinical applications to better understand the molecular mechanisms.

4.2. Histone Alterations in AKI

Histone alterations during translation have the ability to control aging and activate or repress gene expression [124,138]. The types of histone modifications include methylation, acetylation, phosphorylation, ubiquitination, ADP ribosylation, and other histone alterations [124]. Particularly, histone methylation and acetylation are among the most studied epigenetic mechanisms in AKI and repair [124,139]. Global aging-related alterations in H3K9me3, H4K20me3, H3K27me3, and H3K9ac levels have been also documented in both in vitro and in vivo studies [124,140].

4.3. Non-Coding RNA Regulation in AKI

The bulk of the human genome is translated into non-coding RNAs, with less than 2% of it being composed of RNAs having the ability to code for proteins [139]. Non-coding RNAs (ncRNAs) are the genome transcripts that function at the RNA level instead of the protein level, which mainly include microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), small nucleolar RNAs (snoRNAs), and transfer RNAs (tRNAs) [141,142,143,144]. Non-coding RNAs have become significant epigenetic regulators of AKI development and subsequent kidney repair, especially miRNAs and lncRNAs [12,139,145] (Figure 4). Furthermore, non-coding RNAs in circulation may serve as possible biomarkers for AKI [139].

4.3.1. Long Non-Coding RNAs (lncRNA) and AKI

Long non-coding RNAs (lncRNAs) are >200 bp long non-protein-encoding RNAs, which were discovered earlier than miRNA [12,146]. Brannan et al. found the first lncRNA, H19, in 1990, and then a series of lncRNAs was identified [12,147,148]. lncRNAs can attach to histone-modifying complexes to decrease the expression of genes at both the transcriptional and post-transcriptional stages [12,143,149,150]. Nevertheless, there are not enough direct data to draw a conclusion on the role of lncRNAs in kidney healing after AKI; moreover, there is growing proof that lncRNAs play a role in kidney illnesses such as AKI, CKD, kidney transplantation, renal cell cancer, and others [12,139,151].
Using RNA-seq whole-transcriptome profiling reveals that three lncRNAs (MIR210HG, long intergenic non-coding (linc)-ATP13A4–8, and linc-KIAA1737–2) were found in human kidney biopsy samples from kidney transplant patients and were significantly stimulated by hypoxia and cytokines; nevertheless, it is unknown what role these lncRNAs play in AKI and healing [152,153,154].
A different study that employed chromatin immunoprecipitation (ChIP) sequencing of HIF1 and RNA-seq revealed that DARS antisense RNA1 (DARS-AS1) silencing promoted death in cultured renal tubular cells in response to hypoxia, suggesting that DARS-AS1 guards against cell damage brought on by hypoxia [139,153,155].
RANTES, an essential inflammatory mediator in ischemic AKI, is regulated upon the activation of the normal T-cell. The regulation of RANTES synthesis under hypoxic settings by lncRNA-PRINS raises the possibility that lncRNA-PRINS plays a role in the pathophysiology of AKI [139,155].
According to a 2015 study, MALAT1 was the most highly elevated lncRNA in the kidneys of mice that were exposed to inspiratory hypoxia, especially in the proximal tubular cells [139,156]. A more recent study on plasma samples from patients with AKI and biopsy samples from humans with ischemic injury showed the MALAT1 levels to be increased [139,157]. When taken as a whole, these results suggest that lncRNAs might be essential for controlling AKI.

4.3.2. AKI and miRNAs

MicroRNAs, also known as non-coding single-stranded endogenous miRNAs, have between 19 and 23 nucleotides in length and are involved in post-transcriptional gene silencing in all eukaryotes [158,159,160,161,162,163]. Ambros’s group in 1993 first discovered miRNA in Caenorhabditis elegans and showed surprisingly high conservation across species [158,162,163,164]. The biogenesis of miRNAs starts in the nucleus, where the miRNA gene is transcribed by RNA polymerase II, to produce a long primary miRNA [165,166,167] (Figure 5).
A number of conditions, such as ischemia, toxins, sepsis, the obstruction of the urinary tract or the bladder’s outflow, and inflammation, may cause AKI. The dramatic protective effect seen in proximal tubule-specific Dicer knockout mice, where over 80% of miRNAs were reduced, provided the first proof of the crucial function of miRNA in AKI [168]. It is of note that although miRNAs have been reported to play a variety of roles in the kidney, including cellular homeostasis, apoptosis, collagen deposition, and tubular function in I/R-induced AKI, their roles in young vs. old kidney vasculature are still a matter of conjecture. Nevertheless, AKI is commonly seen in the older population. However, the research involving aging animals in this area is limited compared to young animals [169,170,171,172,173,174].
Small non-coding RNAs are known as microRNAs (miRNAs) that control more than 90% of the genes in our body by post-transcriptional regulation. Studies have demonstrated that miRNAs are required for podocyte [175], tubular function [176,177], and renin expression by the juxtaglomerular cells [178].
Wei et al. reported the first instance of miRNAs playing pathogenic roles in AKI and developed a Dicer-knockout mouse model [168]. Proximal tubular cells were precisely eliminated from the Dicer gene, which is required to produce miRNAs. In the renal cortex of these mice, there is a global downregulation of microRNAs. They have normal histology and renal function under controlled conditions but show resistance to the AKI that follows bilateral renal I/R. In comparison to their wild-type counterparts, Dicer-null mice exhibit markedly improved renal function, fewer instances of tissue damage, less tubular apoptosis, and higher survival rates under the latter circumstances [168]. These findings therefore suggest a net pathogenic role for microRNA expression in ischemic AKI. Overall, more than 50 individual microRNAs have now been described as having differential expression in AKI [162].
Many of these microRNAs are involved in experimental and clinical AKI and have been associated with pathways at the molecular level that cause inflammation, apoptosis, and fibrosis. The anti-inflammatory, anti-apoptotic, anti-fibrotic, and pro-angiogenic activities of other AKI-associated microRNAs, on the other hand, might operate as protective mechanisms. Therefore, there are a number of crucial factors to consider when evaluating microRNA patterns in AKI [179].
The miRNA-21 is expressed in the kidneys of mammals and, among others, in heart and lung tissue [180]. In numerous preclinical and clinical I/R research, it has been analyzed and assessed as an important player in tissue protection. For example, it reduces renal I/R damage by preventing apoptosis [181]. Suppressing miRNA-21 expression leads to decreased TNF activity and monocyte chemo-attractant protein-1 (MCP-1), and thereby kidney inflammation [182]. Contrarily, a number of studies have shown that miRNA-21 is differentially released in plasma and/or urine in AKI and is pro-inflammatory. It is often increased in AKI and promotes apoptosis, inflammation, and fibrosis in the kidney [183,184]. Thus, miRNA-21 appears to have a dual function; while it inhibits apoptosis and inflammation to protect against injury, it may also amplify the injury response and encourage fibrosis. According to studies, miRNA-21 prevents apoptosis by upregulating B-cell lymphoma 2 (BCL-2), downregulating the phosphatase and tensin homolog (PTEN), activating the AKT pathway, and downregulating the programmed cell death protein 4 (PDCD4) [185]. It also lowers the levels of the active caspase-3 and caspase-8 proteins [181,186]. The upregulation of miRNA-21 also reduces inflammation by increasing IL-10 and lowering the expression of NF-kB, TNF, interleukin 6 (IL-6), and IL-18 [181]. In animal models of AKI, the experimental upregulation of miRNA-21 offers morphologic and functional renoprotection [181,186,187]. In addition, evidence suggests that the overexpression of miRNA-21 suppresses CSE mRNA and protein in cultured aortic smooth muscle cells and injured carotid arteries, which in turn reduces H2S production [188]. Thus, its role on tissue, i.e., whether protective or damaging, appears to be pathology- and severity-dependent. Table 1 summarizes the findings indicating miRNA-21 as a potential biomarker of AKI in animal models and human samples, and Table 2 summarizes other miRNAs that are associated with AKI.
Table 1. The miRNA-21 studies that are focused on biomarker potential in AKI in animal models and human samples.
Table 1. The miRNA-21 studies that are focused on biomarker potential in AKI in animal models and human samples.
miRNA-21 as Potential Biomarker in Animal Models of AKI
Sample Type and StatusAKI ModelOrigin of SampleRefs.
miRNA-21 was upregulated in kidneys after warm ischemia in miceUnilateral renal ischemia in miceKidney tissue[189]
Urinary miRNA-21 is a marker of hypertensive kidney injury in ratsHypertensive mice with renal tubular lesionUrine[190]
miRNA-21 and the miRNA-17-family are activated after I/R injury in miceLethal and sub-lethal ischemia in miceKidney tissue[191]
miRNA-21 upregulated in rat kidneys after I/R injuryI/R injury or gentamicin in ratsPlasma and urine[192]
miRNA-21 was increased on day 8 after dosingCisplatin-induced kidney injury in rat Urine/Kidney tissue [193]
miRNA-21 was increased in AKI preceding the increase in blood urea nitrogen and creatinineAristolochic acid I (AAI)-induced AKI in rat Plasma[194]
miRNA-21 as Potential Biomarker in Patients with AKI in Human
AKI Model/PopulationSample TypeRole in AKIRefs.
Patients with AKI (n = 98; 27 kidney transplant recipients with biopsy-proven tubular damage and 71 AKI patients in the intensive care unit (ICU)) and patients without AKI (n = 97; 74 healthy volunteers and 23 ICU patients without AKI).UrineBiomarker of diagnosis[195]
120 adult patients undergoing cardiac surgery: 40 non-AKI controls, 39 patients with progressive AKI, and 41 with non-progressive AKI.Urine and plasmaBiomarker of diagnosis[196]
Consecutive patients (n = 115) undergoing major cardiac surgery. Plasma Biomarker of diagnosis[197]
miRNA-10a is renal tubule-specific and is released from tissues upon injury [162]. Its levels in urine and kidney tissue are elevated and lowered in mouse models of renal I/R injury and streptozocin (STZ)-induced diabetic nephropathy (DN), respectively [198,199]. It is believed that miRNA-10a targets IL-12/IL-23p40 to exert protective effects during damage and a pro-apoptotic BCL2-family protein (BIM) [200]. Among critically ill patients in intensive care units (ICUs), reduced plasma levels of miRNA-10a have been demonstrated to predict AKI [201].
In cisplatin nephrotoxic AKI, to prevent damage and death to tubular cells, it is reported that p53 induces miRNA-34a [202]. The miRNA-107 belongs to the miRNA-15/107 family. Members of this family share the AGCAGC at their 5′ end and are involved in a number of pathways that are important for AKI, such as angiogenesis and the stress response [203]. Additionally, miRNA-107 was discovered to be upregulated in circulating endothelial cells in patients with septic-induced AKI [204].
A study has shown that miRNA-129 is involved in Ang-II-induced renal inflammation through an epigenetic mechanism [205]. In 10–12 weeks old mice undergoing unilateral warm ischemia, global miRNA profiling revealed the upregulation of miRNA-21 and miRNA-20a on day one, and on day three, revealed the upregulation of miRNA-146, -199a-3p, and -214 [189]. Other downregulated miRNAs include miRNAs-192, -194, and -197 [189]. The inhibition of miRNA-24 has been reported to suppress the apoptosis of epithelial and endothelial cells, thereby increasing vascular density and reducing tubular fibrosis [206]. These reports suggest that miRNAs play a great role in I/R-induced AKI; however, the dysregulation of miRNAs and their functional relevance in aging kidneys have yet to be fully deciphered. Although miRNA-194 is differentially expressed in I/R-injured kidneys, its role in H2S production remains unknown [189].
The dysregulation of several miRNAs has also been reported in renal diseases, including miRNA-192 involving collagen deposition in DN [207], and the upregulation of miRNAs-155, -452, -224, and -210 in renal cell carcinoma [208]. Additionally, inhibiting miRNA-181 has been shown to cause Bcl-2 to be upregulated and Bax to be downregulated, protecting proximal tubular cells from damage from cisplatin [209].
The transforming growth factor-β (TGF-β)-induced miRNA-214 has been shown to upregulate the AKT pathway, inhibit monocyte and macrophage apoptosis, and promote renal interstitial fibrosis [210,211], whereas a report suggested that the function of miRNA-21 in promoting fibrosis in renal injury is independent of TGF-β signaling [212]. Nevertheless, miRNA-214 is upregulated in various models of AKI and renal fibrosis as well as in the monocytes of animals with CKD [212,213,214]. Renal fibrosis has been shown to be improved by the experimental antagonism of miRNA-214 [212]. Increased urinary miRNA-494 was associated with AKI, and this rise preceded changes in serum creatinine and urea nitrogen [215]. Since there were no variations in miRNA-494 serum concentrations, it was hypothesized that the miRNA-494 was of renal or urinary tract origin [215]. The same investigators then used a murine model of I/R injury to demonstrate an upregulation of miRNA-494 following injury, which inhibited the expression of activating transcription factor 3 (ATF3, a stress-response, reno-protective protein) and increased IL-6, MCP-1, and p-selectin [215]. In addition, miRNA-494 upregulated the expression of inflammatory cytokines and adhesion molecules through a nuclear factor-kappa B (NF-κB)-dependent pathway after kidney I/R injury. The same group discovered that miRNA-494 targeted numerous genes, including adiponectin receptor 2 (ADIPOR2), the B-cell lymphoma 2-like 11 apoptosis facilitator, and the IGF1 receptor (IGF1R), in the human Ingenuity Pathway Analysis or Target Scan System, in addition to ATF3. This would increase inflammation and cause more damage [215]. Mice receiving antisense-miRNA-494 before ischemia miRNA-494 had a clear pathophysiological effect in this AKI model because it ameliorated renal injury, as evaluated by serum creatinine, decreased proinflammatory cytokines, and caused the inhibition of caspase-3 apoptotic activity. Increased urinary miRNA-494 levels are reflective of AKI [215].
Bhatt et. al. showed that during renal I/R in mice and hypoxia in cultured kidney cells, miRNA-687 was noticeably elevated in the kidney [216]. They found that HIF-1 activated miRNA-687, which increased apoptosis by suppressing the phosphatase and tensin homolog (PTEN) [216]. Their animal studies have demonstrated that blocking miRNA-687 prevented kidney damage by maintaining PTEN expression, attenuated cell cycle activation, and reduced apoptosis, collectively suggesting that the HIF-1/miR-687/PTEN signaling pathway in I/R injury may be targeted for therapy [216].
Table 2. The miRNAs that are associated with AKI.
Table 2. The miRNAs that are associated with AKI.
miRNASamples TypeSpeciesAKI Model/PopulationRole in AKIReference
miRNA as biomarkers
miRNA-10aPlasmaRatI/R-induced kidney injury 12 h after reperfusionBiomarker of diagnosis[198]
SerumHumanIntensive care unit (ICU) and cardiac surgery (CS) patientsBiomarker of diagnosis[201]
miRNA-23aSerumHumanSepsis-induced acute kidney injury (AKI) (n = 6), sepsis-non-AKI (n = 6), and healthy volunteers (n = 3)Biomarker of diagnosis[217]
SerumHumanAcute myocardial infarction (AMI) AKI patientsBiomarker of diagnosis[218]
miRNA-30 familyPlasma/Kidney tissueHuman/RatContrast-induced nephropathy (CIN)Biomarker of diagnosis[219]
PlasmaHuman/RatContrast-induced acute kidney injury (CI-AKI)Biomarker of diagnosis[220]
UrineHuman/RatI/R-induced kidney injuryBiomarker of diagnosis[221]
miRNA-126SerumHumanIntensive care units (ICU) and cardiac surgery (CS) patientsBiomarker of diagnosis[201]
miRNA-127SerumHumanIntensive care units (ICU) and cardiac surgery (CS) patientsBiomarker of diagnosis[201]
miRNA-146UrineRatNephrotoxicity study in rats (cisplatin-induced kidney injury)Biomarker of diagnosis[193]
SerumHumanIntensive care units (ICU) and cardiac surgery (CS) patientsBiomarker of diagnosis[201]
miRNA-192UrineRatNephrotoxicity study in rats (cisplatin-induced kidney injury)Biomarker of diagnosis[193]
UrineHuman/RatI/R-induced kidney injuryBiomarker of diagnosis[221]
miRNA-210PlasmaHumanCritically ill patients with acute kidney injury (AKI)Biomarker of diagnosis[222]
SerumHumanIntensive care units (ICU) and cardiac surgery (CS) patientsBiomarker of diagnosis[201]
miRNA-494Urine and kidney tissueHuman/MouseI/R-induced kidney injuryBiomarker of diagnosis[215]
miRNA-489UrineRatGentamicin-induced kidney injuryBiomarker of diagnosis[223]
miRNA-668Urine, serum, rat proximal tubular cells, kidney tissueHuman/MouseI/R-induced kidney injuryBiomarker of diagnosis[224]

5. AKI, Inflammation, and H2S

Hydrogen sulfide (H2S), similar to nitric oxide and carbon monoxide, is a gasotransmitter. It is implicated in a number of inflammatory and vascular disorders that are associated with both pro- and anti-inflammatory signaling, including reducing the adhesion and rolling of circulating leukocytes in inflamed microvasculature, accelerating the resolution of experimental colitis, and enhancing gastric ulcer healing [102,225,226,227,228,229]. H2S can also modulate the immune response in the kidney, reduce inflammation, and promote tissue repair [230,231]. Additionally, it increases blood flow to the kidneys, aiding in their protection against injury [232,233]. Since then, it has been documented that a high amount of NaHS (1 mM) causes TNF-α and IL-1 release from IFN-γ primed U937 cells in a NF-kB-dependent fashion [234]. CSE, an enzyme that produces H2S, was found to be upregulated by LPS in a concentration-dependent manner in a study using primary macrophages (Mϕ) [235]. Conversely, H2S donors have been reported to reduce LPS-induced TNF-α release by microglia [21,27,236] and NF-kβ activation in RAW 264.7 macrophages [28]. Notably, mice deficient with the CBS gene suffer severe growth retardation and mortality within 5 weeks of birth [237]. Mice with CSE deficiency exhibit a milder phenotype, including age-related hypertension and sex-related hyperhomocysteinemia [238].
Numerous factors can lead to AKI and are categorized into three main groups: prerenal, caused by reduced kidney perfusion; renal, with obvious intrinsic kidney damage; and postrenal, caused by a blockage in the urinary tract [239]. However, renal I/R injury represents a leading cause of AKI [240]. I/R injury is a pathological condition that manifests in two steps: first, the blood supply to an organ is restricted, and then second, perfusion and re-oxygenation are restored [241]. One of the most vulnerable organs to I/R injury is the kidney [242]. Renal I/R is followed by a robust inflammatory response in which macrophages play a key role as mediators of inflammation and repair [243]. The two different functional subsets of macrophages are reported—classically activated macrophages (M1), which are mainly linked to pro-inflammatory immune responses, and alternatively activated macrophages (M2), which are primarily involved in tissue repair—and are used to classify macrophages, whether they are recruited or tissue-resident [244]. In an in vitro study using macrophages, lipopolysaccharide (LPS) stimulation decreased H2S production significantly [245]. Similarly, in an in vivo study on zymosan-induced peritonitis, granulocyte infiltration was associated with reduced H2S production [245]. When macrophages were treated with a slow-releasing H2S donor, GYY4137, it was found to suppress inflammatory cytokines and to promote anti-inflammatory chemokines in a concentration-dependent manner [246].
Resident renal mononuclear phagocytes may release proinflammatory cytokines, such as TNF-α, IL-1, and IL-6, and chemokines, such as CCL2, CCL5, CXCL10, and CXCL2, during renal I/R injury [247,248,249,250]. Therefore, a limited percentage of circulating Ly6C-monocytes and Ly6C+ monocytes invade the damaged kidney through a CCL2/CCR2 signaling cascade [247,249,251]. One hour after reperfusion, the wounded kidney experiences an increase in the influx of macrophages, which peaks at 24 h and lasts for 7 days [247]. The accumulation of macrophages in the postischemic kidney’s outer medulla is a hallmark of ongoing inflammation and tissue repair [252]. Infiltrating Ly6C+ monocytes may develop into classically activated macrophages, i.e., M1 macrophages, which express a proinflammatory phenotype in response to damage-associated molecular patterns (DAMPs) or proinflammatory mediators [240,247,252]. Exposure to lipopolysaccharide (LPS), IFN-γ, TNF-α, or GM-CSF induces M1 macrophages [252,253,254]. Neighboring immune cells, such as neutrophils, NK cells, and Th1/Th17 cells, release these inflammatory mediators in the renal interstitium [252]. Then, in a positive feedback loop, M1 macrophages release proinflammatory cytokines (such as TNF-α, IL-1, and IL-6) and ROS, which further increase I/R injury-induced AKI. Indeed, M1 macrophages play a role in both the activation of neutrophils and the triggering of apoptosis in epithelial cells [252,253,255]. Inducible nitric oxide synthase 2 (iNOS), IL-12, IL-23, and Ly6C are all highly expressed on these M1 macrophages, making them easy to spot [253,256]. The classical activation of the macrophage (M1) is shown on the left side of Figure 6.
M1 macrophages exhibit a proinflammatory phenotype with potent antimicrobial activity, and they stimulate or intensify CD4+ T cell polarization to the Th1 state by releasing IL-12 [257]. It is interesting to note that the liposomal clodronate (LC) suppression of kidney macrophages at the earliest stages of I/R injury lowers AKI and enhances renal healing, suggesting a crucial role for macrophages in I/R injury-induced AKI. The harmful function of M1 macrophages in ischemia AKI is further supported by the adoptive transfer of IFN-stimulated macrophages in LC-treated I/R injury mice, which worsens AKI [249,251,253,258,259,260,261,262,263]. Following the initial stages of I/R injury, Th2 and regulatory T (Tregs) cells are enlisted in the damaged renal tissue and produce significant amounts of IL-4, IL-10, and IL-13 [8,252,253,260,264,265,266,267,268,269].
In response to exposure to Th2-type cytokines (IL-4 and IL-13), macrophages change to the anti-inflammatory M2 phenotype, also known as alternatively activated macrophages, which is characterized by high Arg1 (arginase-1) expression, the mannose receptor (MR, also known as CD206), the chitinase-like protein (e.g., Ym1), the resistin-like protein (Fizz1), CD36 (fatty acid translocase), and IL-10 associated with the downregulated expression of pro-inflammatory markers, i.e., IL-12 and iNOS [8,252,253,254,255,257,267,268,269]. Notably, M2 macrophages can develop either directly from invading monocytes or through a transition from the M1 to M2 phenotype [253]. A tissue milieu that would encourage macrophage polarization towards the M2 profile is also created by the macrophage uptake of apoptotic cells that release large levels of anti-inflammatory cytokines, such as TGF-β and IL-10, in conjunction with a decrease in DAMPs [270,271,272]. An alternative activation of the macrophage (M2) is shown on the right side of Figure 6.
M2 macrophages exhibit an anti-inflammatory character and are essential for the immune response against parasites, wound healing, and fibrosis [273,274]. The three distinct subsets of M2 macrophages are M2a, which is triggered by exposure to IL-4 or IL-13, M2b, which is triggered by immune complexes like LPS or IL-1, and M2c, which is triggered by IL-10, TGF-β, or glucocorticoids. While M2c macrophages actively take part in tissue remodeling and exhibit regulatory features, M2a and M2b macrophages support a Th2 immune response, and although they have been identified in vitro, the subsets of macrophages do not accurately represent their activity in vivo [252,257]. These three distinct subgroups are recognized by distinctive gene signatures, which shed light on their role in the pathophysiology of I/R injury-induced AKI [275].
The opposing functions have been attributed to two subsets of macrophages, where M1 is pro-inflammatory and M2 is a regulator of tissue repair [244]. We have shown that H2S expedited macrophage differentiation from M1 to M2 in AKI, both in the young and old kidneys with a greater extent in the young kidney [232]. The above information suggests an intricate mechanism of AKI and repair process involving macrophages in the inflammatory and anti-inflammatory response, and H2S is a regulator of that process. It is therefore important to evaluate H2S production and availability in the kidney under pathological conditions, such as in I/R for effective diagnosis and therapeutic strategies, particularly in aging AKI.

6. AKI and Matrix Biology: Role of EMMPRIN, Meprin-A, and MMPs

The extracellular matrix metalloproteinase inducer (EMMPRIN), also known as CD147 or Basigin, belongs to the immunoglobulin (Ig) superfamily. It is abundant on the surface of tumor cells and stimulates the production of several matrix metalloproteinases (MMPs) by adjacent stromal cells. It is also widely expressed in numerous cell types [154,276,277]. It acts as a cell surface receptor for a variety of ligands to activate a number of MMPs, which are involved in a number of processes, such as cell disruption, inflammation, tissue healing, and remodeling [277].
Notably, MMPs are a family of enzymes that can degrade a wide variety of extracellular protein (ECM) proteins [278]. There are over 20 different MMPs, and they are classified into four groups: collagenases, gelatinases, stromelysins, and membrane-type MMPs [278]. Anti-EMMPRIN antibody therapy is cardioprotective, while EMMPRIN-induced MMP-9 production has been shown to promote myocardial I/R injury [279].
Meprins are astacin family zinc-dependent metalloproteinases that were first discovered and characterized in the mouse’s brush-border membranes [280], rat kidneys [281], and human intestines [282]. In addition to EMMPRIN, metallo-endopeptidase Meprin-A also activates MMP-9 [283]. Within the kidney, Meprin-A localizes to the membranes of the brush-border of proximal tubules [284]. Numerous studies have shown that Meprin-A plays a significant part in AKI, which involves ECM degradation leading to remodeling [285,286]. Others have shown that elevated MMP-9 expression and activity cause extensive damage to the tubules, glomeruli, and interstitial regions [287,288,289]. These studies suggest that the induction of EMMPRIN, Meprin-A, and MMP-9 is intricately associated with AKI in the young. However, the extent of their dysregulation in I/R-induced AKI in aging was not clear until we recently showed that EMMPRIN, Meprin-A, and MMP-9 are also upregulated in aging AKI [232]. The extent of their upregulation was higher than young AKI, and H2S mitigated their expression, both in young and old kidneys, suggesting that H2S plays a crucial role in their regulation during AKI pathology. Additionally, it has been reported that hyperhomocysteinemia (HHcy), which causes a decrease in H2S levels, increases the M1 macrophage phenotype, and this effect was partially attributed to EMMPRIN induction [290].
MMPs represent inflammatory cell-derived cytokines, another important group of fibrosis-related inflammatory cytokines [291,292,293]. MMP-2 and -9 were both associated with the progression of renal fibrosis [291,293,294]. MMPs suppressed fibrosis through the degradation of ECM components [292,295]. In NaHS treatment, the renal expression of MMP-2 and MMP-9 in diabetic kidney disease rats was downregulated [293,296,297]. Our results indicated that diabetic kidneys express high levels of MMP-9 with increased activity, resulting in the downregulation of H2S production by diminishing CBS and CSE enzymes. In the downstream pathway, low levels of H2S induced NMDA-R1 and elevated Cxs-40 and -43. When considered collectively, these findings imply that H2S plays a major role as a mediator in diabetic kidney remodeling [298]. It has also been suggested that GYY4137 therapy stabilizes MMP-9, MMP-13, and MMP-14 levels and lowers ROS-induced kidney fibrosis by increasing microRNA-194 [299].
In addition to MMP-9, other MMPs such as MMP-1, -2, -3, -8, and -13 are also related to ECM remodeling during progressive renal diseases [300,301]. Collagens I and III are degraded by the interstitial collagenases MMP-1, -8, and -13 [302], whereas MMP-3 favors ECM proteins, particularly fibronectin [303]. The role of MMP-7 in the progression of tubulointerstitial fibrosis is well known [304], and MMP-12 in podocytes is implicated in glomerulonephritis [305]. Interestingly, MMP activity is regulated by the tissue inhibitors of metalloproteinases (TIMPs), and TIMP-1 inactivates most MMPs [306,307,308].
So, we can conclude that H2S has the therapeutic potential to reduce the remodeling of the kidneys associated with diabetes while also suppressing the inflammatory response. Further research is still needed to fully understand how H2S modulation affects the inflammation of the AKI.

7. ECM and Vascular Dysfunction in Renal Aging and AKI

The endothelium extensively covers the inner surface of the cardiovascular system. Its polarized architecture in blood vessels, heterogeneity in morphology, structure, and gene expression profile at various locations of different blood vessel types demonstrate its functional importance [309]. Vascular inflammation normally begins and spreads in the endothelium. H2S suppresses vascular inflammation [102,309] by activating KATP and BKCa channels as well as HO-1 expression, inhibiting p38, and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) [310,311]. Additionally, H2S reduces the amount of reactive oxygen species (ROS) in endothelial cells [62,312,313,314], which is accomplished in part by scavenging ROS [105,315] and in part by improving antioxidant defense mechanisms.
I/R injury is defined as cellular damage by ischemia followed by reperfusion of formerly viable ischemic tissues [316]. Endothelial injury is the hallmark of I/R injury [317]. Ischemia promotes the expression of pro-inflammatory molecules and cytokines in the endothelium while decreasing the production of beneficial molecules, including nitric oxide, thrombomodulin, and prostacyclin [316]. In addition, it may decrease H2S production as there is evidence that CBS, CSE, and 3-MST are also present in the vascular endothelium [23,318,319,320,321].
Pathological situations in the adult organism, such as injury, inflammation, or aging, can activate endothelial-to-mesenchymal transition (EndoMT) and cause the fibrosis of the affected organs [322]. In addition to renal fibrosis, the EndoMT has been postulated to play a role in the initiation and advancement of cardiac, pulmonary, hepatic, corneal, intestinal, unilateral ureteral obstruction (UUO), diabetes, and wound healing [323,324,325,326,327].
In a groundbreaking study, Zeisberg et al. used three mouse models of renal fibrosis—UUO, a model for studying progressive tubulointerstitial fibrosis, streptozotocin (STZ)-induced DN, and alpha 3 chain of collagen type 4 (COL4A3) knockout mice—to confirm first the role of EndoMT in renal fibrosis [327]. A considerable fraction of myofibroblasts co-expressed the endothelium marker CD31, also referred to as platelet endothelial cell adhesion molecule-1 (PECAM-1), in all three models. Additionally, they co-expressed the markers for myofibroblasts, α-smooth muscle actin (α-SMA), and fibroblast-specific protein-1 (FSP-1, equivalent to S100A4) [327]. During the onset and development of pathological fibrosis, an excessive and uncontrolled production of ECM is caused by the accumulation of many myofibroblasts [323]. Emerging evidence also suggests that the vascular endothelium is one of the sources of myofibroblasts. Endothelial cells become myofibroblasts when tissue-infiltrating chronic inflammatory cells, such as macrophages and lymphocytes, produce TGF-β. The endothelial-derived mesenchymal cells then go into the interstitium and take part in tissue fibrosis [323]. By examining phosphorylated Smad3, the TGF-β pathway’s activation was determined, and TGF-β blocking experiments made use of a particular Smad3 inhibitor (SIS3). These findings showed that Smad3 activation was necessary for advanced glycation end-products (AGEs) to activate EndoMT in microvascular endothelial cells in vitro and in transgenic mice in vivo [323,328]. Scientific evidence also shows that EndoMT is a unique mechanism that contributes to the formation of fibrosis in DN, and it has been proposed that blocking EndoMT using inhibitors of the TGF-β pathway may offer a new strategy to slow the evolution of DN and other fibrotic processes [323]. Lineage tracing indicated that about 30–50% of kidney fibroblasts in UUO nephropathy, DN, and Alport renal disease models expressed endothelial markers, indicating a large contribution of endothelial cells to fibroblasts through phenotypic change [327].
Collagen IV is found in the glomerular basement membrane (GBM), which serves as a structural and functional barrier between the vascular system and the urinary space [329]. Further, the components of the renal interstitial matrix are collagen types I, III, V, VI, VII, and XV, and collagen types I and III are reported to be deposited in the early stages of renal fibrosis [330,331,332].
In an article, Patschan et. al. postulated that mid-term prognosis in AKI depends on, in part, the presence of interstitial fibrosis by EndoMT. They also found that treatment with early endothelial progenitor cells (eEPCs) can reduce postischemic EndoMT and fibrosis in young AKI mice [333]. Additionally, we have demonstrated that during I/R injury, the aging kidney showed increased EndoMT and fibrosis compared to the young kidney [232]. After receiving treatment with GYY4137, an H2S donor, the aging kidney’s vascular density, blood flow, and renal function were all enhanced. We concluded that H2S can rescue kidney dysfunction in aging mice due to the I/R injury that is related to EndoMT and fibrosis [232]. Nevertheless, further and broader studies are needed to confirm these initial findings for a viable approach for aged patients sustaining AKI.

8. Current Treatment Modalities and Future Strategies

The therapeutic strategies for AKI based on the KDIGO guidelines and bundles of care are limited and mostly supportive [334]. There is not any approved drug that has been designated to treat, prevent, or speed up the healing process from AKI. The primary goal of current interventions is to stop additional declines in renal function [335]. The clinical approach should begin with hemodynamic stabilization, the early detection of AKI consequences, understanding of its etiology, and management [336]. Because autoregulation mechanisms are compromised in AKI, hemodynamic stability is crucial [337].
To prevent negative effects, special attention should be given to drugs with renal toxicity, which should be either stopped or the dose adjusted based on renal function [336,338]. Furthermore, it is essential to start antibiotics as soon as possible in septic patients [339].
Finally, when treating an AKI patient, it is critical to quickly detect and address additional issues, such as hyperkalemia, metabolic acidosis, anemia, and fluid overload [334]. It is also advised to begin stress-ulcer prophylaxis and prevent infection during AKI [334]. Figure 7 summarizes current therapeutic modalities in AKI.

8.1. Fluid Therapy

In individuals with AKI, intravascular hypovolemia is a serious consequence. It is corrected by individualized fluid therapy, whose main aim is to increase intravascular blood flow and increase cardiac output [337]. Fluids of fluid therapy are broadly classified into colloid (albumin, starch) and crystalloid (saline, Ringer’s lactate, or PlasmaLyte) [337,340].
Fluid therapy should be personalized based on the patient’s physiological parameters, underlying diagnosis, trajectories, and the overall risks and benefits of fluids [337]. Although the ideal fluid type is still uncertain, starches should be avoided because trials have reported that starches can worsen the incidence of AKI due to osmotic nephrosis with proximal vacuolization and swelling [341].

8.2. Vasopressor Medicine

The median blood pressure of an AKI patient must be greater than 65 mmHg, which can maintain accurate renal perfusion. After fluid therapy or volume resuscitation, a vasopressor drug should be given to ensure enough renal perfusion to prevent a positive fluid balance [334,342,343,344]. The recommended first-line vasopressor in a vasodilatory state is noradrenaline [345,346]. It increases perfusion pressure and maintains circulatory flow. The lowest dose of noradrenaline is recommended because higher doses can cause decreased blood flow through excessive vasoconstriction [347].
Vasopressin and Terlipressin are two effective alternatives for rising blood pressure, although their comparison with noradrenaline has not been demonstrated yet [348,349,350,351]. Angiotensin II has shown promising results on patient outcomes in recent studies, namely, by improving survival and renal function recovery [352]. However, further research is still needed before angiotensin II should be used regularly.

8.3. Diuretics

To achieve electrolyte balance and manage fluid overload in AKI, diuretics are used [334,353]. It was previously thought that loop diuretics might protect the loop of Henle from ischemia by decreasing its workload [353]. It has never been proven that furosemide has any clinical advantages in preventing AKI, reducing the requirement for renal replacement therapy (RRT), promoting renal recovery, or lowering in-hospital mortality [354,355,356]. It is important to note that certain studies have associated the usage of loop diuretics with a higher risk of death, which might be related to the delay in appropriate RRT start [353,356,357]. The disadvantages related to diuretics include how high doses of loop diuretics can cause ototoxicity [353]. As a consequence, the KDIGO guideline does not recommend the use of diuretics as an AKI treatment [334].

8.4. Drug-Induced AKI

Drug toxicity is the major cause of AKI and has been associated with 20–40% of AKI cases, and it goes up to 60% in elderly patients [358,359]. Drug nephrotoxicity is diverse and varies with many classes of drugs. Drugs can cause direct kidney damage, such as tubular injury, interstitial nephritis, glomerular injury, obstructive nephropathy, and indirect harm by reducing renal blood flow [360]. Moreover, drug-induced nephrotoxicity arises from innate drug toxicity, changed renal hemodynamic characters, prior renal issues, and altered drug pharmacokinetics in critical illness [360,361]. Medications and chemicals commonly linked to nephrotoxicity include calcineurin inhibitors, which increase the chance of developing prerenal AKI, renin–angiotensin–aldosterone system blockers, high-dose systemic vasoconstrictors, and nonsteroidal anti-inflammatory medicines (NSAIDs) [358]. Aminoglycosides, vancomycin, radiocontrast agents, cisplatin, amphotericin B, foscarnet, and osmotically active substances are associated with acute tubular necrosis (ATN), which is dose-dependent and the common form of drug-induced AKI in hospitalized patients [358,362]. Patients treated with acyclovir, ciprofloxacin, methotrexate, and sulfa-based medicines may develop AKI as a result of crystal-induced luminal blockage [362].
To reduce toxicity, medication prescriptions must be properly thought out. The KDIGO recommendations urge early drug withdrawal of potentially nephrotoxic medications, avoiding radiocontrast and other potentially harmful pharmaceuticals, and drug dose monitoring [334].

8.5. Renal Replacement Therapy

Anuria, severe/refractory metabolic acidosis, severe/refractory hyperkalemia, severe azotemia, refractory volume overload, or clinical effects of uremia such as encephalopathy, pericarditis, or neuropathy are conventional criteria for initiating RRT in AKI [363,364]. RRT is necessary to preserve the homeostasis of volume, electrolytes, acid-base, and uremic solutes in AKI patients. Additionally, it is hypothesized that RRT can reduce inflammation, which may be important for septic patients; however, this is still unknown [363,365]. The disadvantages of this therapy are that RRT necessitates the insertion of a central venous dialysis catheter, the contact of blood with an extracorporeal circuit, and anticoagulation. It can also be linked to hemodynamic instability, which may delay kidney recovery [363,365]. Although there has not been any proof that the risk classification of patients for AKI can help with long-term renal outcomes, it can enable the use of appropriate treatments and reduce the incidence of AKI [366,367,368].
Perna et. al. showed that the transcriptional dysregulation of genes encoding H2S-producing enzymes resulted in a decrease in H2S levels in hemodialysis patients [369]. In a separate communication, they also reported interesting findings on H2S levels before and after single standard 4 h hemodialysis sessions in 131 patients, which were conducted in two dialysis sessions separated by one month [370]. Their results indicated that plasma H2S was significantly increased and homocysteine (Hcy) levels were significantly decreased after dialysis compared to before dialysis, suggesting that hemodialysis is effective in reestablishing the altered H2S homeostasis in these patients. They also interpreted through reaction stoichiometry analysis that an improved Hcy metabolism occurred through H2S formation during dialysis [370]. These results strongly suggest that RRT is effective in maintaining H2S homeostasis in kidney failure patients, which can possibly be applied to AKI patients as well.

9. Summary and Discussion

Acute kidney injury (AKI) is very common and significantly reduces the normal function of the kidney. The mortality of AKI remains high due to the lack of early diagnosis and cause-specific treatment. Thus, it is a significant burden for both the patient and society. A specific treatment has not yet been developed for AKI probably due to the complex nature of the condition and our inability to recognize AKI phenotypes at this time.
I/R injury is one of the main causes of AKI. AKI prevention and early detection are our main options for lowering the incidence and harmful effects of AKI because there are no viable treatment therapies for established AKI and because of its high morbidity and mortality. Conversely, one could counter that risk assessment is ineffective since it is not evident which interventions are beneficial to use and which ones should be applied to high-risk individuals.
H2S has beneficial effects on aging in the body by acting as an antioxidant against oxidative species such as ROS and RNS. H2S is generated by several enzymes, i.e., CBS, CSE, CAT, and 3MST, and protects cells from damage, reduces inflammation, and improves mitochondrial function. However, more research is needed to confirm these findings and to determine how H2S could be used to slow down the aging process in humans.
Following renal I/R injury, the miRNA expression profile was studied, and it shows a survival response. In fact, the majority of miRNAs that were discovered seem to have differential expression, possibly suppressing the damage response by controlling apoptosis, proliferation, and inflammation. These findings may help us identify novel pathways that can be targeted to prevent or reduce renal injury. If the miRNAs found to be expressed differently can be shown to play a role in these processes in vivo, they may represent new biomarkers of renal injury.
Leukocyte adhesion to the endothelium and endothelial permeability are both regulated by H S. The anti-inflammatory H2S effects seem to be mediated through activating KATP channels; thus, these KATP channels are potential targets for anti-inflammatory therapies in AKI among aged patients. The early influx of macrophages promotes a proinflammatory state that amplifies tissue injuries. In response, M1-recruited monocytes or tissue-resident macrophages change into M2 macrophages in response to local cues, which help to promote tissue repair and inhibit the renal inflammatory response. The depletion of macrophages 3 days after I/R injury slows the rebuilding of renal tissue, but the depletion of macrophages before I/R injury lessens renal insult. More specifically, the information that is currently available strongly suggests that H2S may develop into the next effective preventative and therapeutic agent for preventing and treating aging-related illnesses and age-associated diseases, and this should be studied in different aged AKI models.

10. Future Perspective

The management of AKI is poor in underdeveloped countries due to high costs and late diagnosis. In addition, currently available therapies have major side effects. Nevertheless, a range of specific treatment strategies is emerging that may have an influence on the prognosis of patients with AKI, thanks to recent advancements in the knowledge of its pathophysiology and advancements in trial design. Additional biomarkers could be used to define AKI more precisely, leading to earlier identification [371,372]. While the research on H2S and aging is still in its early stages with knowledge gaps, it is a promising area of research. If H2S has beneficial effects on renal aging, it could potentially be used to develop new therapies for delaying the onset of age-related diseases and extending lifespans. On the other hand, current information demonstrates that miRNAs have a potential role in I/R injury-induced AKI; however, the dysregulation of miRNAs and their functional relevance in aging kidneys have yet to be fully deciphered.

Author Contributions

Conceptualization, U.S. and S.G.; analyses and assembly, S.G., S.M., K.B., S.B.P., and H.A.; manuscript daft, S.G.; supervision, manuscript review, editing, and final draft, U.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Acknowledgments

This work was supported, in part, by the NIH grant DK116591 to U.S. The funder had no role in the study design, management, collection, analysis, and interpretation of data, as well as the writing of the report for publication.

Conflicts of Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Abbreviations

3-MST, 3-mercaptopyruvate sulfur transferase; AKI, acute kidney injury; AKT, protein kinase B; ATF3, activating transcription factor 3; ATN, acute tubular necrosis; BIM, a pro-apoptotic BCL2-family protein; CBS, cystathionine β-synthase; CKD, chronic kidney disease; CSE, cystathionine γ-lyase; DAO, D-amino acid oxidase; DAMPs, damage-associated molecular patterns; DN, diabetic nephropathy; ECM, extracellular protein; EMMPRIN, extracellular matrix metalloproteinase inducer; EndoMT, endothelial-to-mesenchymal transition; GBM, glomerular basement membrane; GFR, glomerular filtration rate; H2S, hydrogen sulfide; I/R, ischemia-reperfusion; IL, interleukin; miRNA, micro RNA; MCP-1, monocyte chemoattractant protein-1; PTEN, phosphatase and tensin homolog; RBF, renal blood flow; ROS, reactive oxygen species; RRT, renal replacement therapy; STZ, streptozotocin; TECs, tubular epithelial cells; TGF-β, transforming growth factor-β; TNF, tumor necrosis factor; TST, thiosulfate transferase; UUO, unilateral ureteral obstruction.

References

  1. Section 2: AKI Definition. Kidney Int. Suppl. (2011) 2012, 2, 19–36.
  2. 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]
  3. Bouchard, J.; Mehta, R.L. Acute Kidney Injury in Western Countries. Kidney Dis. 2016, 2, 103–110. [Google Scholar]
  4. Heung, M.; Yessayan, L. Renal Replacement Therapy in Acute Kidney Injury: Controversies and Consensus. Crit. Care Clin. 2017, 33, 365–378. [Google Scholar] [PubMed]
  5. Hoste, E.A.; 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]
  6. Chawla, L.S.; Bellomo, R.; Bihorac, A.; Goldstein, S.L.; Siew, E.D.; Bagshaw, S.M.; Bittleman, D.; Cruz, D.; Endre, Z.; Fitzgerald, R.L.; et al. Acute kidney disease and renal recovery: Consensus report of the Acute Disease Quality Initiative (ADQI) 16 Workgroup. Nat. Rev. Nephrol. 2017, 13, 241–257. [Google Scholar]
  7. Zhou, L.; Zhuo, H.; Ouyang, H.; Liu, Y.; Yuan, F.; Sun, L.; Liu, F.; Liu, H. Glycoprotein non-metastatic melanoma protein b (Gpnmb) is highly expressed in macrophages of acute injured kidney and promotes M2 macrophages polarization. Cell Immunol. 2017, 316, 53–60. [Google Scholar]
  8. Malek, M.; Nematbakhsh, M. Renal ischemia/reperfusion injury; from pathophysiology to treatment. J. Ren. Inj. Prev. 2015, 4, 20–27. [Google Scholar]
  9. Rodriguez, F.; Bonacasa, B.; Fenoy, F.J.; Salom, M.G. Reactive oxygen and nitrogen species in the renal ischemia/reperfusion injury. Curr. Pharm. Des. 2013, 19, 2776–2794. [Google Scholar]
  10. Chertow, G.M.; Burdick, E.; Honour, M.; Bonventre, J.V.; Bates, D.W. Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. J. Am. Soc. Nephrol. 2005, 16, 3365–3370. [Google Scholar]
  11. Hao, J.; Wei, Q.; Mei, S.; Li, L.; Su, Y.; Mei, C.; Dong, Z. Induction of microRNA-17-5p by p53 protects against renal ischemia-reperfusion injury by targeting death receptor 6. Kidney Int. 2017, 91, 106–118. [Google Scholar]
  12. Liu, Z.; Wang, Y.; Shu, S.; Cai, J.; Tang, C.; Dong, Z. Non-coding RNAs in kidney injury and repair. Am. J. Physiol. Cell Physiol. 2019, 317, C177–C188. [Google Scholar]
  13. Selman, M.; Pardo, A. Fibroageing: An ageing pathological feature driven by dysregulated extracellular matrix-cell mechanobiology. Ageing Res. Rev. 2021, 70, 101393. [Google Scholar] [PubMed]
  14. James, M.T.; Hemmelgarn, B.R.; Wiebe, N.; Pannu, N.; Manns, B.J.; Klarenbach, S.W.; Tonelli, M.; Alberta Kidney Disease, N. Glomerular filtration rate, proteinuria, and the incidence and consequences of acute kidney injury: A cohort study. Lancet 2010, 376, 2096–2103. [Google Scholar] [PubMed]
  15. Sturmlechner, I.; Durik, M.; Sieben, C.J.; Baker, D.J.; van Deursen, J.M. Cellular senescence in renal ageing and disease. Nat. Rev. Nephrol. 2017, 13, 77–89. [Google Scholar] [PubMed]
  16. Docherty, M.H.; O’Sullivan, E.D.; Bonventre, J.V.; Ferenbach, D.A. Cellular Senescence in the Kidney. J. Am. Soc. Nephrol. 2019, 30, 726–736. [Google Scholar]
  17. Zhang, J.Q.; Li, Y.Y.; Zhang, X.Y.; Tian, Z.H.; Liu, C.; Wang, S.T.; Zhang, F.R. Cellular senescence of renal tubular epithelial cells in renal fibrosis. Front. Endocrinol. 2023, 14, 1085605. [Google Scholar]
  18. Rex, N.; Melk, A.; Schmitt, R. Cellular senescence and kidney aging. Clin. Sci. 2023, 137, 1805–1821. [Google Scholar]
  19. Duarte, I.; Gameiro, J.; Resina, C.; Outerelo, C. In-hospital mortality in elderly patients with acute kidney injury requiring dialysis: A cohort analysis. Int. Urol. Nephrol. 2020, 52, 1117–1124. [Google Scholar]
  20. Wang, R. Gasotransmitters: Growing pains and joys. Trends Biochem. Sci. 2014, 39, 227–232. [Google Scholar]
  21. Fagone, P.; Mazzon, E.; Bramanti, P.; Bendtzen, K.; Nicoletti, F. Gasotransmitters and the immune system: Mode of action and novel therapeutic targets. Eur. J. Pharmacol. 2018, 834, 92–102. [Google Scholar] [PubMed]
  22. Mustafa, A.K.; Sikka, G.; Gazi, S.K.; Steppan, J.; Jung, S.M.; Bhunia, A.K.; Barodka, V.M.; Gazi, F.K.; Barrow, R.K.; Wang, R.; et al. Hydrogen sulfide as endothelium-derived hyperpolarizing factor sulfhydrates potassium channels. Circ. Res. 2011, 109, 1259–1268. [Google Scholar] [PubMed]
  23. Yang, G.; Wu, L.; Jiang, B.; Yang, W.; Qi, J.; Cao, K.; Meng, Q.; Mustafa, A.K.; Mu, W.; Zhang, S.; et al. H2S as a physiologic vasorelaxant: Hypertension in mice with deletion of cystathionine gamma-lyase. Science 2008, 322, 587–590. [Google Scholar] [PubMed]
  24. Liu, C.; Gu, X.; Zhu, Y.Z. Synthesis and biological evaluation of novel leonurine-SPRC conjugate as cardioprotective agents. Bioorganic Med. Chem. Lett. 2010, 20, 6942–6946. [Google Scholar]
  25. Taniguchi, S.; Kang, L.; Kimura, T.; Niki, I. Hydrogen sulphide protects mouse pancreatic beta-cells from cell death induced by oxidative stress, but not by endoplasmic reticulum stress. Br. J. Pharmacol. 2011, 162, 1171–1178. [Google Scholar]
  26. Zhang, H.; Zhang, A.; Guo, C.; Shi, C.; Zhang, Y.; Liu, Q.; Sparatore, A.; Wang, C. S-diclofenac protects against doxorubicin-induced cardiomyopathy in mice via ameliorating cardiac gap junction remodeling. PLoS ONE 2011, 6, e26441. [Google Scholar]
  27. Hu, L.F.; Wong, P.T.; Moore, P.K.; Bian, J.S. Hydrogen sulfide attenuates lipopolysaccharide-induced inflammation by inhibition of p38 mitogen-activated protein kinase in microglia. J. Neurochem. 2007, 100, 1121–1128. [Google Scholar]
  28. Oh, G.S.; Pae, H.O.; Lee, B.S.; Kim, B.N.; Kim, J.M.; Kim, H.R.; Jeon, S.B.; Jeon, W.K.; Chae, H.J.; Chung, H.T. Hydrogen sulfide inhibits nitric oxide production and nuclear factor-kappaB via heme oxygenase-1 expression in RAW264.7 macrophages stimulated with lipopolysaccharide. Free Radic. Biol. Med. 2006, 41, 106–119. [Google Scholar]
  29. Sen, N.; Paul, B.D.; Gadalla, M.M.; Mustafa, A.K.; Sen, T.; Xu, R.; Kim, S.; Snyder, S.H. Hydrogen sulfide-linked sulfhydration of NF-kappaB mediates its antiapoptotic actions. Mol. Cell 2012, 45, 13–24. [Google Scholar]
  30. Cohen, H.Y.; Miller, C.; Bitterman, K.J.; Wall, N.R.; Hekking, B.; Kessler, B.; Howitz, K.T.; Gorospe, M.; de Cabo, R.; Sinclair, D.A. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 2004, 305, 390–392. [Google Scholar]
  31. Kim, K.M.; Pae, H.O.; Zhung, M.; Ha, H.Y.; Ha, Y.A.; Chai, K.Y.; Cheong, Y.K.; Kim, J.M.; Chung, H.T. Involvement of anti-inflammatory heme oxygenase-1 in the inhibitory effect of curcumin on the expression of pro-inflammatory inducible nitric oxide synthase in RAW264.7 macrophages. Biomed. Pharmacother. 2008, 62, 630–636. [Google Scholar] [PubMed]
  32. Pan, L.L.; Liu, X.H.; Gong, Q.H.; Wu, D.; Zhu, Y.Z. Hydrogen sulfide attenuated tumor necrosis factor-alpha-induced inflammatory signaling and dysfunction in vascular endothelial cells. PLoS ONE 2011, 6, e19766. [Google Scholar]
  33. Feng, J.; Lu, X.; Li, H.; Wang, S. The roles of hydrogen sulfide in renal physiology and disease states. Ren. Fail. 2022, 44, 1289–1308. [Google Scholar]
  34. Xia, M.; Chen, L.; Muh, R.W.; Li, P.L.; Li, N. Production and actions of hydrogen sulfide, a novel gaseous bioactive substance, in the kidneys. J. Pharmacol. Exp. Ther. 2009, 329, 1056–1062. [Google Scholar] [PubMed]
  35. Ge, S.N.; Zhao, M.M.; Wu, D.D.; Chen, Y.; Wang, Y.; Zhu, J.H.; Cai, W.J.; Zhu, Y.Z.; Zhu, Y.C. Hydrogen sulfide targets EGFR Cys797/Cys798 residues to induce Na(+)/K(+)-ATPase endocytosis and inhibition in renal tubular epithelial cells and increase sodium excretion in chronic salt-loaded rats. Antioxid. Redox Signal. 2014, 21, 2061–2082. [Google Scholar]
  36. Dugbartey, G.J. Physiological role of hydrogen sulfide in the kidney and its therapeutic implications for kidney diseases. Biomed. Pharmacother. 2023, 166, 115396. [Google Scholar]
  37. Mathai, J.C.; Missner, A.; Kugler, P.; Saparov, S.M.; Zeidel, M.L.; Lee, J.K.; Pohl, P. No facilitator required for membrane transport of hydrogen sulfide. Proc. Natl. Acad. Sci. USA 2009, 106, 16633–16638. [Google Scholar]
  38. Sun, H.J.; Wu, Z.Y.; Cao, L.; Zhu, M.Y.; Liu, T.T.; Guo, L.; Lin, Y.; Nie, X.W.; Bian, J.S. Hydrogen Sulfide: Recent Progression and Perspectives for the Treatment of Diabetic Nephropathy. Molecules 2019, 24, 2857. [Google Scholar] [CrossRef]
  39. Cao, X.; Bian, J.S. The Role of Hydrogen Sulfide in Renal System. Front. Pharmacol. 2016, 7, 385. [Google Scholar]
  40. Kashfi, K. The role of hydrogen sulfide in health and disease. Biochem. Pharmacol. 2018, 149, 1–4. [Google Scholar]
  41. Lee, H.J.; Feliers, D.; Barnes, J.L.; Oh, S.; Choudhury, G.G.; Diaz, V.; Galvan, V.; Strong, R.; Nelson, J.; Salmon, A.; et al. Hydrogen sulfide ameliorates aging-associated changes in the kidney. GeroScience 2018, 40, 163–176. [Google Scholar] [PubMed]
  42. Jin, H.F.; Liang, C.; Liang, J.M.; Tang, C.S.; Du, J.B. Effects of hydrogen sulfide on vascular inflammation in pulmonary hypertension induced by high pulmonary blood flow: Experiment with rats. Zhonghua Yi Xue Za Zhi 2008, 88, 2235–2239. [Google Scholar] [PubMed]
  43. Abe, K.; Kimura, H. The possible role of hydrogen sulfide as an endogenous neuromodulator. J. Neurosci. 1996, 16, 1066–1071. [Google Scholar] [PubMed]
  44. Hosoki, R.; Matsuki, N.; Kimura, H. The possible role of hydrogen sulfide as an endogenous smooth muscle relaxant in synergy with nitric oxide. Biochem. Biophys. Res. Commun. 1997, 237, 527–531. [Google Scholar] [PubMed]
  45. Liu, Y.H.; Yan, C.D.; Bian, J.S. Hydrogen sulfide: A novel signaling molecule in the vascular system. J. Cardiovasc. Pharmacol. 2011, 58, 560–569. [Google Scholar]
  46. Lowicka, E.; Beltowski, J. Hydrogen sulfide (H2S)—The third gas of interest for pharmacologists. Pharmacol. Rep. 2007, 59, 4–24. [Google Scholar]
  47. Gai, J.W.; Wahafu, W.; Guo, H.; Liu, M.; Wang, X.C.; Xiao, Y.X.; Zhang, L.; Xin, Z.C.; Jin, J. Further evidence of endogenous hydrogen sulphide as a mediator of relaxation in human and rat bladder. Asian J. Androl. 2013, 15, 692–696. [Google Scholar]
  48. Srilatha, B.; Adaikan, P.G.; Li, L.; Moore, P.K. Hydrogen sulphide: A novel endogenous gasotransmitter facilitates erectile function. J. Sex. Med. 2007, 4, 1304–1311. [Google Scholar]
  49. Braunstein, A.E.; Goryachenkova, E.V.; Tolosa, E.A.; Willhardt, I.H.; Yefremova, L.L. Specificity and some other properties of liver serine sulphhydrase: Evidence for its identity with cystathionine -synthase. Biochim. Biophys. Acta 1971, 242, 247–260. [Google Scholar]
  50. Chiku, T.; Padovani, D.; Zhu, W.; Singh, S.; Vitvitsky, V.; Banerjee, R. H2S biogenesis by human cystathionine gamma-lyase leads to the novel sulfur metabolites lanthionine and homolanthionine and is responsive to the grade of hyperhomocysteinemia. J. Biol. Chem. 2009, 284, 11601–11612. [Google Scholar]
  51. Feng, S.; Ji, J.; Li, H.; Zhang, X. H(2)S alleviates renal ischemia and reperfusion injury by suppressing ERS-induced autophagy. Transpl. Immunol. 2024, 83, 102006. [Google Scholar] [PubMed]
  52. Shibuya, N.; Mikami, Y.; Kimura, Y.; Nagahara, N.; Kimura, H. Vascular endothelium expresses 3-mercaptopyruvate sulfurtransferase and produces hydrogen sulfide. J. Biochem. 2009, 146, 623–626. [Google Scholar] [PubMed]
  53. Whiteman, M.; Le Trionnaire, S.; Chopra, M.; Fox, B.; Whatmore, J. Emerging role of hydrogen sulfide in health and disease: Critical appraisal of biomarkers and pharmacological tools. Clin. Sci. 2011, 121, 459–488. [Google Scholar]
  54. Kimura, H. Hydrogen Sulfide (H(2)S)/Polysulfides (H(2)S(n)) Signalling and TRPA1 Channels Modification on Sulfur Metabolism. Biomolecules 2024, 14, 129. [Google Scholar] [CrossRef]
  55. Feliers, D.; Lee, H.J.; Kasinath, B.S. Hydrogen Sulfide in Renal Physiology and Disease. Antioxid. Redox Signal. 2016, 25, 720–731. [Google Scholar]
  56. Szlezak, D.; Hutsch, T.; Ufnal, M.; Wrobel, M. Heart and kidney H(2)S production is reduced in hypertensive and older rats. Biochimie 2022, 199, 130–138. [Google Scholar] [PubMed]
  57. Correia, M.J.; Pimpao, A.B.; Fernandes, D.G.F.; Morello, J.; Sequeira, C.O.; Calado, J.; Antunes, A.M.M.; Almeida, M.S.; Branco, P.; Monteiro, E.C.; et al. Cysteine as a Multifaceted Player in Kidney, the Cysteine-Related Thiolome and Its Implications for Precision Medicine. Molecules 2022, 27, 1416. [Google Scholar] [CrossRef]
  58. Stipanuk, M.H.; Beck, P.W. Characterization of the enzymic capacity for cysteine desulphhydration in liver and kidney of the rat. Biochem. J. 1982, 206, 267–277. [Google Scholar] [PubMed]
  59. Mikami, Y.; Shibuya, N.; Kimura, Y.; Nagahara, N.; Ogasawara, Y.; Kimura, H. Thioredoxin and dihydrolipoic acid are required for 3-mercaptopyruvate sulfurtransferase to produce hydrogen sulfide. Biochem. J. 2011, 439, 479–485. [Google Scholar]
  60. House, J.D.; Brosnan, M.E.; Brosnan, J.T. Characterization of homocysteine metabolism in the rat kidney. Biochem. J. 1997, 328 Pt 1, 287–292. [Google Scholar]
  61. Yuan, X.; Zhang, J.; Xie, F.; Tan, W.; Wang, S.; Huang, L.; Tao, L.; Xing, Q.; Yuan, Q. Loss of the Protein Cystathionine beta-Synthase During Kidney Injury Promotes Renal Tubulointerstitial Fibrosis. Kidney Blood Press. Res. 2017, 42, 428–443. [Google Scholar] [PubMed]
  62. Bos, E.M.; Wang, R.; Snijder, P.M.; Boersema, M.; Damman, J.; Fu, M.; Moser, J.; Hillebrands, J.L.; Ploeg, R.J.; Yang, G.; et al. Cystathionine gamma-lyase protects against renal ischemia/reperfusion by modulating oxidative stress. J. Am. Soc. Nephrol. 2013, 24, 759–770. [Google Scholar] [PubMed]
  63. Nagahara, N.; Ito, T.; Kitamura, H.; Nishino, T. Tissue and subcellular distribution of mercaptopyruvate sulfurtransferase in the rat: Confocal laser fluorescence and immunoelectron microscopic studies combined with biochemical analysis. Histochem. Cell Biol. 1998, 110, 243–250. [Google Scholar] [PubMed]
  64. Stipanuk, M.H.; Ueki, I. Dealing with methionine/homocysteine sulfur: Cysteine metabolism to taurine and inorganic sulfur. J. Inherit. Metab. Dis. 2011, 34, 17–32. [Google Scholar]
  65. Kasinath, B.S.; Feliers, D.; Lee, H.J. Hydrogen sulfide as a regulatory factor in kidney health and disease. Biochem. Pharmacol. 2018, 149, 29–41. [Google Scholar]
  66. Paul, B.D.; Snyder, S.H. H(2)S signalling through protein sulfhydration and beyond. Nat. Rev. Mol. Cell Biol. 2012, 13, 499–507. [Google Scholar]
  67. Modis, K.; Coletta, C.; Erdelyi, K.; Papapetropoulos, A.; Szabo, C. Intramitochondrial hydrogen sulfide production by 3-mercaptopyruvate sulfurtransferase maintains mitochondrial electron flow and supports cellular bioenergetics. FASEB J. 2013, 27, 601–611. [Google Scholar]
  68. Shibuya, N.; Koike, S.; Tanaka, M.; Ishigami-Yuasa, M.; Kimura, Y.; Ogasawara, Y.; Fukui, K.; Nagahara, N.; Kimura, H. A novel pathway for the production of hydrogen sulfide from D-cysteine in mammalian cells. Nat. Commun. 2013, 4, 1366. [Google Scholar]
  69. Yang, M.; Zhou, Y.; Wang, K.; Luo, C.; Xie, M.; Shi, X.; Lin, X. Review of Chemical Sensors for Hydrogen Sulfide Detection in Organisms and Living Cells. Sensors 2023, 23, 3316. [Google Scholar] [CrossRef]
  70. Zhang, X.; Zhou, W.; Yuan, Z.; Lu, C. Colorimetric detection of biological hydrogen sulfide using fluorosurfactant functionalized gold nanorods. Analyst 2015, 140, 7443–7450. [Google Scholar]
  71. Yuan, Z.; Lu, F.; Peng, M.; Wang, C.W.; Tseng, Y.T.; Du, Y.; Cai, N.; Lien, C.W.; Chang, H.T.; He, Y.; et al. Selective Colorimetric Detection of Hydrogen Sulfide Based on Primary Amine-Active Ester Cross-Linking of Gold Nanoparticles. Anal. Chem. 2015, 87, 7267–7273. [Google Scholar] [PubMed]
  72. Jia, T.T.; Zhang, Y.; Hou, J.T.; Niu, H.; Wang, S. H(2)S-based fluorescent imaging for pathophysiological processes. Front. Chem. 2023, 11, 1126309. [Google Scholar]
  73. Lin, V.S.; Chen, W.; Xian, M.; Chang, C.J. Chemical probes for molecular imaging and detection of hydrogen sulfide and reactive sulfur species in biological systems. Chem. Soc. Rev. 2015, 44, 4596–4618. [Google Scholar]
  74. Yao, L.; Zhang, W.; Yin, C.; Zhang, Y.; Huo, F. A tracer-type fluorescent probe for imaging adenosine triphosphate under the stresses of hydrogen sulfide and hydrogen peroxide in living cells. Analyst 2022, 147, 4222–4227. [Google Scholar] [PubMed]
  75. Nagarkar, S.S.; Saha, T.; Desai, A.V.; Talukdar, P.; Ghosh, S.K. Metal-organic framework based highly selective fluorescence turn-on probe for hydrogen sulphide. Sci. Rep. 2014, 4, 7053. [Google Scholar]
  76. Sun, Y.; Sun, P.; Li, Z.; Qu, L.; Guo, W. Natural flavylium-inspired far-red to NIR-II dyes and their applications as fluorescent probes for biomedical sensing. Chem. Soc. Rev. 2022, 51, 7170–7205. [Google Scholar] [PubMed]
  77. Qi, Y.L.; Wang, H.R.; Chen, L.L.; Duan, Y.T.; Yang, S.Y.; Zhu, H.L. Recent advances in small-molecule fluorescent probes for studying ferroptosis. Chem. Soc. Rev. 2022, 51, 7752–7778. [Google Scholar]
  78. Hanaoka, K.; Iwaki, S.; Yagi, K.; Myochin, T.; Ikeno, T.; Ohno, H.; Sasaki, E.; Komatsu, T.; Ueno, T.; Uchigashima, M.; et al. General Design Strategy to Precisely Control the Emission of Fluorophores via a Twisted Intramolecular Charge Transfer (TICT) Process. J. Am. Chem. Soc. 2022, 144, 19778–19790. [Google Scholar]
  79. Zhang, H.; Zhao, H.; Guo, N. Protective effect of hydrogen sulfide on the kidney (Review). Mol. Med. Rep. 2021, 24, 696. [Google Scholar]
  80. Mustafa, A.K.; Gadalla, M.M.; Sen, N.; Kim, S.; Mu, W.; Gazi, S.K.; Barrow, R.K.; Yang, G.; Wang, R.; Snyder, S.H. H2S signals through protein S-sulfhydration. Sci. Signal. 2009, 2, ra72. [Google Scholar]
  81. Kabil, O.; Banerjee, R. Redox biochemistry of hydrogen sulfide. J. Biol. Chem. 2010, 285, 21903–21907. [Google Scholar]
  82. Fu, M.; Zhang, W.; Wu, L.; Yang, G.; Li, H.; Wang, R. Hydrogen sulfide (H2S) metabolism in mitochondria and its regulatory role in energy production. Proc. Natl. Acad. Sci. USA 2012, 109, 2943–2948. [Google Scholar] [PubMed]
  83. Roy, A.; Khan, A.H.; Islam, M.T.; Prieto, M.C.; Majid, D.S. Interdependency of cystathione gamma-lyase and cystathione beta-synthase in hydrogen sulfide-induced blood pressure regulation in rats. Am. J. Hypertens. 2012, 25, 74–81. [Google Scholar] [PubMed]
  84. Luo, R.; Hu, S.; Liu, Q.; Han, M.; Wang, F.; Qiu, M.; Li, S.; Li, X.; Yang, T.; Fu, X.; et al. Hydrogen sulfide upregulates renal AQP-2 protein expression and promotes urine concentration. FASEB J. 2019, 33, 469–483. [Google Scholar]
  85. Zhao, W.; Wang, R. H(2)S-induced vasorelaxation and underlying cellular and molecular mechanisms. Am. J. Physiol. Heart Circ. Physiol. 2002, 283, H474–H480. [Google Scholar] [PubMed]
  86. Olson, K.R. Hydrogen sulfide as an oxygen sensor. Antioxid. Redox Signal. 2015, 22, 377–397. [Google Scholar]
  87. Koning, A.M.; Frenay, A.R.; Leuvenink, H.G.; van Goor, H. Hydrogen sulfide in renal physiology, disease and transplantation--the smell of renal protection. Nitric Oxide 2015, 46, 37–49. [Google Scholar] [PubMed]
  88. Beltowski, J. Hypoxia in the renal medulla: Implications for hydrogen sulfide signaling. J. Pharmacol. Exp. Ther. 2010, 334, 358–363. [Google Scholar]
  89. Olson, K.R.; Dombkowski, R.A.; Russell, M.J.; Doellman, M.M.; Head, S.K.; Whitfield, N.L.; Madden, J.A. Hydrogen sulfide as an oxygen sensor/transducer in vertebrate hypoxic vasoconstriction and hypoxic vasodilation. J. Exp. Biol. 2006, 209 Pt 20, 4011–4023. [Google Scholar]
  90. Olson, K.R. Hydrogen sulfide as an oxygen sensor. Clin. Chem. Lab. Med. 2013, 51, 623–632. [Google Scholar]
  91. Prieto-Lloret, J.; Aaronson, P.I. Hydrogen Sulfide as an O(2) Sensor: A Critical Analysis. Adv. Exp. Med. Biol. 2017, 967, 261–276. [Google Scholar] [PubMed]
  92. Teng, H.; Wu, B.; Zhao, K.; Yang, G.; Wu, L.; Wang, R. Oxygen-sensitive mitochondrial accumulation of cystathionine beta-synthase mediated by Lon protease. Proc. Natl. Acad. Sci. USA 2013, 110, 12679–12684. [Google Scholar]
  93. Peters, J.; Munter, K.; Bader, M.; Hackenthal, E.; Mullins, J.J.; Ganten, D. Increased adrenal renin in transgenic hypertensive rats, TGR(mREN2)27, and its regulation by cAMP, angiotensin II, and calcium. J. Clin. Investig. 1993, 91, 742–747. [Google Scholar]
  94. Schweda, F.; Friis, U.; Wagner, C.; Skott, O.; Kurtz, A. Renin release. Physiology 2007, 22, 310–319. [Google Scholar] [PubMed]
  95. Liu, J.; Zhou, Y.; Liu, Y.; Li, L.; Chen, Y.; Liu, Y.; Feng, Y.; Yosypiv, I.V.; Song, R.; Peng, H. (Pro)renin receptor regulates lung development via the Wnt/beta-catenin signaling pathway. Am. J. Physiol. Lung Cell Mol. Physiol. 2019, 317, L202–L211. [Google Scholar] [PubMed]
  96. Yang, T.; Xu, C. Physiology and Pathophysiology of the Intrarenal Renin-Angiotensin System: An Update. J. Am. Soc. Nephrol. 2017, 28, 1040–1049. [Google Scholar]
  97. Fountain, J.H.; Kaur, J.; Lappin, S.L. Physiology, Renin Angiotensin System. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2024. [Google Scholar]
  98. Lu, M.; Liu, Y.H.; Goh, H.S.; Wang, J.J.; Yong, Q.C.; Wang, R.; Bian, J.S. Hydrogen sulfide inhibits plasma renin activity. J. Am. Soc. Nephrol. 2010, 21, 993–1002. [Google Scholar]
  99. Laggner, H.; Hermann, M.; Esterbauer, H.; Muellner, M.K.; Exner, M.; Gmeiner, B.M.; Kapiotis, S. The novel gaseous vasorelaxant hydrogen sulfide inhibits angiotensin-converting enzyme activity of endothelial cells. J. Hypertens. 2007, 25, 2100–2104. [Google Scholar]
  100. Szabo, C.; Coletta, C.; Chao, C.; Modis, K.; Szczesny, B.; Papapetropoulos, A.; Hellmich, M.R. Tumor-derived hydrogen sulfide, produced by cystathionine-beta-synthase, stimulates bioenergetics, cell proliferation, and angiogenesis in colon cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 12474–12479. [Google Scholar]
  101. Wilkie, S.E.; Borland, G.; Carter, R.N.; Morton, N.M.; Selman, C. Hydrogen sulfide in ageing, longevity and disease. Biochem. J. 2021, 478, 3485–3504. [Google Scholar]
  102. Zanardo, R.C.; Brancaleone, V.; Distrutti, E.; Fiorucci, S.; Cirino, G.; Wallace, J.L. Hydrogen sulfide is an endogenous modulator of leukocyte-mediated inflammation. FASEB J. 2006, 20, 2118–2120. [Google Scholar] [PubMed]
  103. Lazarevic, M.; Battaglia, G.; Jevtic, B.; Dedovic, N.; Bruno, V.; Cavalli, E.; Miljkovic, D.; Nicoletti, F.; Momcilovic, M.; Fagone, P. Upregulation of Tolerogenic Pathways by the Hydrogen Sulfide Donor GYY4137 and Impaired Expression of H(2)S-Producing Enzymes in Multiple Sclerosis. Antioxidants 2020, 9, 608. [Google Scholar] [CrossRef] [PubMed]
  104. Gemici, B.; Elsheikh, W.; Feitosa, K.B.; Costa, S.K.; Muscara, M.N.; Wallace, J.L. H2S-releasing drugs: Anti-inflammatory, cytoprotective and chemopreventative potential. Nitric Oxide 2015, 46, 25–31. [Google Scholar] [PubMed]
  105. Tratnig-Frankl, P.; Andrews, A.R.; Berkane, Y.; Guinier, C.; Goutard, M.; Lupon, E.; Lancia, H.H.; Morrison, M.L.; Roth, M.B.; Randolph, M.A.; et al. Exploring Iodide and Hydrogen Sulfide as ROS Scavengers to Delay Acute Rejection in MHC-Defined Vascularized Composite Allografts. Antioxidants 2024, 13, 531. [Google Scholar] [CrossRef]
  106. Ahmad, A.; Sattar, M.Z.; Rathore, H.A.; Hussain, A.I.; Khan, S.A.; Fatima, T.; Afzal, S.; Abdullah, N.A.; Johns, E.J. Antioxidant Activity and Free Radical Scavenging Capacity of L-Arginine and Nahs: A Comparative in Vitro Study. Acta Pol. Pharm. 2015, 72, 245–252. [Google Scholar]
  107. Szabo, C.; Papapetropoulos, A. International Union of Basic and Clinical Pharmacology. CII: Pharmacological Modulation of H(2)S Levels: H(2)S Donors and H(2)S Biosynthesis Inhibitors. Pharmacol. Rev. 2017, 69, 497–564. [Google Scholar] [PubMed]
  108. Vintila, A.R.; Slade, L.; Cooke, M.; Willis, C.R.G.; Torregrossa, R.; Rahman, M.; Anupom, T.; Vanapalli, S.A.; Gaffney, C.J.; Gharahdaghi, N.; et al. Mitochondrial sulfide promotes life span and health span through distinct mechanisms in developing versus adult treated Caenorhabditis elegans. Proc. Natl. Acad. Sci. USA 2023, 120, e2216141120. [Google Scholar]
  109. Castello, L.; Froio, T.; Cavallini, G.; Biasi, F.; Sapino, A.; Leonarduzzi, G.; Bergamini, E.; Poli, G.; Chiarpotto, E. Calorie restriction protects against age-related rat aorta sclerosis. FASEB J. 2005, 19, 1863–1865. [Google Scholar]
  110. Anantharaju, A.; Feller, A.; Chedid, A. Aging Liver: A review. Gerontology 2002, 48, 343–353. [Google Scholar]
  111. Schmitt, R.; Melk, A. Molecular mechanisms of renal aging. Kidney Int. 2017, 92, 569–579. [Google Scholar]
  112. Mani, S.; Li, H.; Untereiner, A.; Wu, L.; Yang, G.; Austin, R.C.; Dickhout, J.G.; Lhotak, S.; Meng, Q.H.; Wang, R. Decreased endogenous production of hydrogen sulfide accelerates atherosclerosis. Circulation 2013, 127, 2523–2534. [Google Scholar] [PubMed]
  113. Albertini, E.; Koziel, R.; Durr, A.; Neuhaus, M.; Jansen-Durr, P. Cystathionine beta synthase modulates senescence of human endothelial cells. Aging 2012, 4, 664–673. [Google Scholar] [PubMed]
  114. Predmore, B.L.; Alendy, M.J.; Ahmed, K.I.; Leeuwenburgh, C.; Julian, D. The hydrogen sulfide signaling system: Changes during aging and the benefits of caloric restriction. Age 2010, 32, 467–481. [Google Scholar]
  115. Susantitaphong, P.; Cruz, D.N.; Cerda, J.; Abulfaraj, M.; Alqahtani, F.; Koulouridis, I.; Jaber, B.L.; Acute Kidney Injury Advisory Group of the American Society of Nephrology. World incidence of AKI: A meta-analysis. Clin. J. Am. Soc. Nephrol. 2013, 8, 1482–1493. [Google Scholar]
  116. Hoste, E.A.J.; Kellum, J.A.; Selby, N.M.; Zarbock, A.; Palevsky, P.M.; Bagshaw, S.M.; Goldstein, S.L.; Cerda, J.; Chawla, L.S. Global epidemiology and outcomes of acute kidney injury. Nat. Rev. Nephrol. 2018, 14, 607–625. [Google Scholar]
  117. Gameiro, J.; Fonseca, J.A.; Outerelo, C.; Lopes, J.A. Acute Kidney Injury: From Diagnosis to Prevention and Treatment Strategies. J. Clin. Med. 2020, 9, 1704. [Google Scholar] [CrossRef] [PubMed]
  118. Bagshaw, S.M.; George, C.; Bellomo, R.; Committee, A.D.M. Changes in the incidence and outcome for early acute kidney injury in a cohort of Australian intensive care units. Crit. Care 2007, 11, R68. [Google Scholar]
  119. Hsu, R.K.; McCulloch, C.E.; Dudley, R.A.; Lo, L.J.; Hsu, C.Y. Temporal changes in incidence of dialysis-requiring AKI. J. Am. Soc. Nephrol. 2013, 24, 37–42. [Google Scholar] [PubMed]
  120. Bellomo, R. The epidemiology of acute renal failure: 1975 versus 2005. Curr. Opin. Crit. Care 2006, 12, 557–560. [Google Scholar]
  121. Cruz, D.N.; Ronco, C. Acute kidney injury in the intensive care unit: Current trends in incidence and outcome. Crit. Care 2007, 11, 149. [Google Scholar]
  122. Wing, M.R.; Ramezani, A.; Gill, H.S.; Devaney, J.M.; Raj, D.S. Epigenetics of progression of chronic kidney disease: Fact or fantasy? Semin. Nephrol. 2013, 33, 363–374. [Google Scholar] [PubMed]
  123. Wanner, N.; Bechtel-Walz, W. Epigenetics of kidney disease. Cell Tissue Res. 2017, 369, 75–92. [Google Scholar] [PubMed]
  124. Wang, K.; Liu, H.; Hu, Q.; Wang, L.; Liu, J.; Zheng, Z.; Zhang, W.; Ren, J.; Zhu, F.; Liu, G.H. Epigenetic regulation of aging: Implications for interventions of aging and diseases. Signal Transduct. Target. Ther. 2022, 7, 374. [Google Scholar] [PubMed]
  125. Jin, B.; Li, Y.; Robertson, K.D. DNA methylation: Superior or subordinate in the epigenetic hierarchy? Genes. Cancer 2011, 2, 607–617. [Google Scholar]
  126. Ramsahoye, B.H.; Biniszkiewicz, D.; Lyko, F.; Clark, V.; Bird, A.P.; Jaenisch, R. Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc. Natl. Acad. Sci. USA 2000, 97, 5237–5242. [Google Scholar] [PubMed]
  127. Lister, R.; Pelizzola, M.; Dowen, R.H.; Hawkins, R.D.; Hon, G.; Tonti-Filippini, J.; Nery, J.R.; Lee, L.; Ye, Z.; Ngo, Q.M.; et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 2009, 462, 315–322. [Google Scholar]
  128. Shirane, K.; Toh, H.; Kobayashi, H.; Miura, F.; Chiba, H.; Ito, T.; Kono, T.; Sasaki, H. Mouse oocyte methylomes at base resolution reveal genome-wide accumulation of non-CpG methylation and role of DNA methyltransferases. PLoS Genet. 2013, 9, e1003439. [Google Scholar]
  129. Guo, J.U.; Su, Y.; Shin, J.H.; Shin, J.; Li, H.; Xie, B.; Zhong, C.; Hu, S.; Le, T.; Fan, G.; et al. Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain. Nat. Neurosci. 2014, 17, 215–222. [Google Scholar]
  130. Xie, W.; Barr, C.L.; Kim, A.; Yue, F.; Lee, A.Y.; Eubanks, J.; Dempster, E.L.; Ren, B. Base-resolution analyses of sequence and parent-of-origin dependent DNA methylation in the mouse genome. Cell 2012, 148, 816–831. [Google Scholar]
  131. Schubeler, D. Function and information content of DNA methylation. Nature 2015, 517, 321–326. [Google Scholar]
  132. Jeltsch, A. Beyond Watson and Crick: DNA methylation and molecular enzymology of DNA methyltransferases. Chembiochem 2002, 3, 274–293. [Google Scholar] [PubMed]
  133. Smith, S.S.; Kaplan, B.E.; Sowers, L.C.; Newman, E.M. Mechanism of human methyl-directed DNA methyltransferase and the fidelity of cytosine methylation. Proc. Natl. Acad. Sci. USA 1992, 89, 4744–4748. [Google Scholar] [PubMed]
  134. Jones, P.L.; Veenstra, G.J.; Wade, P.A.; Vermaak, D.; Kass, S.U.; Landsberger, N.; Strouboulis, J.; Wolffe, A.P. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet. 1998, 19, 187–191. [Google Scholar] [PubMed]
  135. Liu, F.; Chen, J.; Li, Z.; Meng, X. Recent Advances in Epigenetics of Age-Related Kidney Diseases. Genes 2022, 13, 796. [Google Scholar] [CrossRef]
  136. Gao, Q.; Chen, F.; Zhang, L.; Wei, A.; Wang, Y.; Wu, Z.; Cao, W. Inhibition of DNA methyltransferase aberrations reinstates antioxidant aging suppressors and ameliorates renal aging. Aging Cell 2022, 21, e13526. [Google Scholar]
  137. Pushpakumar, S.; Ren, L.; Juin, S.K.; Majumder, S.; Kulkarni, R.; Sen, U. Methylation-dependent antioxidant-redox imbalance regulates hypertensive kidney injury in aging. Redox Biol. 2020, 37, 101754. [Google Scholar]
  138. Kouzarides, T. Chromatin modifications and their function. Cell 2007, 128, 693–705. [Google Scholar]
  139. Guo, C.; Dong, G.; Liang, X.; Dong, Z. Epigenetic regulation in AKI and kidney repair: Mechanisms and therapeutic implications. Nat. Rev. Nephrol. 2019, 15, 220–239. [Google Scholar]
  140. Sidler, C.; Kovalchuk, O.; Kovalchuk, I. Epigenetic Regulation of Cellular Senescence and Aging. Front. Genet. 2017, 8, 138. [Google Scholar]
  141. Bali, K.K.; Kuner, R. Noncoding RNAs: Key molecules in understanding and treating pain. Trends Mol. Med. 2014, 20, 437–448. [Google Scholar]
  142. Kota, S.K.; Kota, S.B. Noncoding RNA and epigenetic gene regulation in renal diseases. Drug Discov. Today 2017, 22, 1112–1122. [Google Scholar] [PubMed]
  143. Kato, M. Noncoding RNAs as therapeutic targets in early stage diabetic kidney disease. Kidney Res. Clin. Pract. 2018, 37, 197–209. [Google Scholar]
  144. Vallone, C.; Rigon, G.; Gulia, C.; Baffa, A.; Votino, R.; Morosetti, G.; Zaami, S.; Briganti, V.; Catania, F.; Gaffi, M.; et al. Non-Coding RNAs and Endometrial Cancer. Genes 2018, 9, 187. [Google Scholar] [CrossRef] [PubMed]
  145. Brandenburger, T.; Salgado Somoza, A.; Devaux, Y.; Lorenzen, J.M. Noncoding RNAs in acute kidney injury. Kidney Int. 2018, 94, 870–881. [Google Scholar] [PubMed]
  146. Liu, X.; Hao, Y.; Yu, W.; Yang, X.; Luo, X.; Zhao, J.; Li, J.; Hu, X.; Li, L. Long Non-Coding RNA Emergence During Renal Cell Carcinoma Tumorigenesis. Cell Physiol. Biochem. 2018, 47, 735–746. [Google Scholar]
  147. Brannan, C.I.; Dees, E.C.; Ingram, R.S.; Tilghman, S.M. The product of the H19 gene may function as an RNA. Mol. Cell Biol. 1990, 10, 28–36. [Google Scholar]
  148. Louro, R.; Smirnova, A.S.; Verjovski-Almeida, S. Long intronic noncoding RNA transcription: Expression noise or expression choice? Genomics 2009, 93, 291–298. [Google Scholar]
  149. Hu, W.; Alvarez-Dominguez, J.R.; Lodish, H.F. Regulation of mammalian cell differentiation by long non-coding RNAs. Embo Rep. 2012, 13, 971–983. [Google Scholar]
  150. Lalevee, S.; Feil, R. Long noncoding RNAs in human disease: Emerging mechanisms and therapeutic strategies. Epigenomics 2015, 7, 877–879. [Google Scholar]
  151. Moghaddas Sani, H.; Hejazian, M.; Hosseinian Khatibi, S.M.; Ardalan, M.; Zununi Vahed, S. Long non-coding RNAs: An essential emerging field in kidney pathogenesis. Biomed. Pharmacother. 2018, 99, 755–765. [Google Scholar]
  152. Lin, J.; Zhang, X.; Xue, C.; Zhang, H.; Shashaty, M.G.; Gosai, S.J.; Meyer, N.; Grazioli, A.; Hinkle, C.; Caughey, J.; et al. The long noncoding RNA landscape in hypoxic and inflammatory renal epithelial injury. Am. J. Physiol. Ren. Physiol. 2015, 309, F901–F913. [Google Scholar]
  153. Mimura, I.; Hirakawa, Y.; Kanki, Y.; Kushida, N.; Nakaki, R.; Suzuki, Y.; Tanaka, T.; Aburatani, H.; Nangaku, M. Novel lnc RNA regulated by HIF-1 inhibits apoptotic cell death in the renal tubular epithelial cells under hypoxia. Physiol. Rep. 2017, 5, e13203. [Google Scholar]
  154. Guo, H.; Li, R.; Zucker, S.; Toole, B.P. EMMPRIN (CD147), an inducer of matrix metalloproteinase synthesis, also binds interstitial collagenase to the tumor cell surface. Cancer Res. 2000, 60, 888–891. [Google Scholar]
  155. Yu, T.M.; Palanisamy, K.; Sun, K.T.; Day, Y.J.; Shu, K.H.; Wang, I.K.; Shyu, W.C.; Chen, P.; Chen, Y.L.; Li, C.Y. RANTES mediates kidney ischemia reperfusion injury through a possible role of HIF-1α and LncRNA PRINS. Sci. Rep. 2016, 6, 18424. [Google Scholar]
  156. Lelli, A.; Nolan, K.A.; Santambrogio, S.; Goncalves, A.F.; Schonenberger, M.J.; Guinot, A.; Frew, I.J.; Marti, H.H.; Hoogewijs, D.; Wenger, R.H. Induction of long noncoding RNA MALAT1 in hypoxic mice. Hypoxia 2015, 3, 45–52. [Google Scholar]
  157. Kolling, M.; Genschel, C.; Kaucsar, T.; Hubner, A.; Rong, S.; Schmitt, R.; Sorensen-Zender, I.; Haddad, G.; Kistler, A.; Seeger, H.; et al. Hypoxia-induced long non-coding RNA Malat1 is dispensable for renal ischemia/reperfusion-injury. Sci. Rep. 2018, 8, 3438. [Google Scholar]
  158. Lee, R.C.; Feinbaum, R.L.; Ambros, V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993, 75, 843–854. [Google Scholar]
  159. Du, T.; Zamore, P.D. microPrimer: The biogenesis and function of microRNA. Development 2005, 132, 4645–4652. [Google Scholar]
  160. Bottinger, E.P. TGF-beta in renal injury and disease. Semin. Nephrol. 2007, 27, 309–320. [Google Scholar]
  161. Wang, G.K.; Zhu, J.Q.; Zhang, J.T.; Li, Q.; Li, Y.; He, J.; Qin, Y.W.; Jing, Q. Circulating microRNA: A novel potential biomarker for early diagnosis of acute myocardial infarction in humans. Eur. Heart J. 2010, 31, 659–666. [Google Scholar]
  162. Fan, P.C.; Chen, C.C.; Chen, Y.C.; Chang, Y.S.; Chu, P.H. MicroRNAs in acute kidney injury. Hum. Genom. 2016, 10, 29. [Google Scholar]
  163. Mukhadi, S.; Hull, R.; Mbita, Z.; Dlamini, Z. The Role of MicroRNAs in Kidney Disease. Noncoding RNA 2015, 1, 192–221. [Google Scholar] [PubMed]
  164. Small, E.M.; Olson, E.N. Pervasive roles of microRNAs in cardiovascular biology. Nature 2011, 469, 336–342. [Google Scholar] [PubMed]
  165. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar]
  166. Macfarlane, L.A.; Murphy, P.R. MicroRNA: Biogenesis, Function and Role in Cancer. Curr. Genom. 2010, 11, 537–561. [Google Scholar]
  167. Liu, J.; Zhou, F.; Guan, Y.; Meng, F.; Zhao, Z.; Su, Q.; Bao, W.; Wang, X.; Zhao, J.; Huo, Z.; et al. The Biogenesis of miRNAs and Their Role in the Development of Amyotrophic Lateral Sclerosis. Cells 2022, 11, 572. [Google Scholar] [CrossRef]
  168. Wei, Q.; Bhatt, K.; He, H.Z.; Mi, Q.S.; Haase, V.H.; Dong, Z. Targeted deletion of Dicer from proximal tubules protects against renal ischemia-reperfusion injury. J. Am. Soc. Nephrol. 2010, 21, 756–761. [Google Scholar]
  169. Mahtal, N.; Lenoir, O.; Tinel, C.; Anglicheau, D.; Tharaux, P.L. MicroRNAs in kidney injury and disease. Nat. Rev. Nephrol. 2022, 18, 643–662. [Google Scholar]
  170. Chandrasekaran, K.; Karolina, D.S.; Sepramaniam, S.; Armugam, A.; Wintour, E.M.; Bertram, J.F.; Jeyaseelan, K. Role of microRNAs in kidney homeostasis and disease. Kidney Int. 2012, 81, 617–627. [Google Scholar]
  171. Wu, Y.L.; Li, H.F.; Chen, H.H.; Lin, H. MicroRNAs as Biomarkers and Therapeutic Targets in Inflammation- and Ischemia-Reperfusion-Related Acute Renal Injury. Int. J. Mol. Sci. 2020, 21, 6738. [Google Scholar] [CrossRef]
  172. Zou, Y.F.; Zhang, W. Role of microRNA in the detection, progression, and intervention of acute kidney injury. Exp. Biol. Med. 2018, 243, 129–136. [Google Scholar]
  173. Guan, H.; Peng, R.; Mao, L.; Fang, F.; Xu, B.; Chen, M. Injured tubular epithelial cells activate fibroblasts to promote kidney fibrosis through miR-150-containing exosomes. Exp. Cell Res. 2020, 392, 112007. [Google Scholar] [PubMed]
  174. Anderson, S.; Eldadah, B.; Halter, J.B.; Hazzard, W.R.; Himmelfarb, J.; Horne, F.M.; Kimmel, P.L.; Molitoris, B.A.; Murthy, M.; O’Hare, A.M.; et al. Acute kidney injury in older adults. J. Am. Soc. Nephrol. 2011, 22, 28–38. [Google Scholar] [PubMed]
  175. Ho, J.; Ng, K.H.; Rosen, S.; Dostal, A.; Gregory, R.I.; Kreidberg, J.A. Podocyte-specific loss of functional microRNAs leads to rapid glomerular and tubular injury. J. Am. Soc. Nephrol. 2008, 19, 2069–2075. [Google Scholar]
  176. Shi, S.; Yu, L.; Chiu, C.; Sun, Y.; Chen, J.; Khitrov, G.; Merkenschlager, M.; Holzman, L.B.; Zhang, W.; Mundel, P.; et al. Podocyte-selective deletion of dicer induces proteinuria and glomerulosclerosis. J. Am. Soc. Nephrol. 2008, 19, 2159–2169. [Google Scholar]
  177. Sepramaniam, S.; Armugam, A.; Lim, K.Y.; Karolina, D.S.; Swaminathan, P.; Tan, J.R.; Jeyaseelan, K. MicroRNA 320a functions as a novel endogenous modulator of aquaporins 1 and 4 as well as a potential therapeutic target in cerebral ischemia. J. Biol. Chem. 2010, 285, 29223–29230. [Google Scholar]
  178. Sequeira-Lopez, M.L.; Weatherford, E.T.; Borges, G.R.; Monteagudo, M.C.; Pentz, E.S.; Harfe, B.D.; Carretero, O.; Sigmund, C.D.; Gomez, R.A. The microRNA-processing enzyme dicer maintains juxtaglomerular cells. J. Am. Soc. Nephrol. 2010, 21, 460–467. [Google Scholar]
  179. Jones, T.F.; Bekele, S.; O’Dwyer, M.J.; Prowle, J.R. MicroRNAs in Acute Kidney Injury. Nephron 2018, 140, 124–128. [Google Scholar]
  180. Lagos-Quintana, M.; Rauhut, R.; Yalcin, A.; Meyer, J.; Lendeckel, W.; Tuschl, T. Identification of tissue-specific microRNAs from mouse. Curr. Biol. 2002, 12, 735–739. [Google Scholar]
  181. Hu, H.; Jiang, W.; Xi, X.; Zou, C.; Ye, Z. MicroRNA-21 attenuates renal ischemia reperfusion injury via targeting caspase signaling in mice. Am. J. Nephrol. 2014, 40, 215–223. [Google Scholar]
  182. Zhong, X.; Chung, A.C.; Chen, H.Y.; Dong, Y.; Meng, X.M.; Li, R.; Yang, W.; Hou, F.F.; Lan, H.Y. miR-21 is a key therapeutic target for renal injury in a mouse model of type 2 diabetes. Diabetologia 2013, 56, 663–674. [Google Scholar] [PubMed]
  183. Chau, B.N.; Xin, C.; Hartner, J.; Ren, S.; Castano, A.P.; Linn, G.; Li, J.; Tran, P.T.; Kaimal, V.; Huang, X.; et al. MicroRNA-21 promotes fibrosis of the kidney by silencing metabolic pathways. Sci. Transl. Med. 2012, 4, 121ra118. [Google Scholar]
  184. Li, Y.F.; Jing, Y.; Hao, J.; Frankfort, N.C.; Zhou, X.; Shen, B.; Liu, X.; Wang, L.; Li, R. MicroRNA-21 in the pathogenesis of acute kidney injury. Protein Cell 2013, 4, 813–819. [Google Scholar] [PubMed]
  185. Shakeri, A.; Gahnbari, M.; Tasbandi, A.; Sahebkar, A. Regulation of microRNA-21 expression by natural products in cancer. Phytother. Res. 2021, 35, 3732–3746. [Google Scholar]
  186. Jia, P.; Teng, J.; Zou, J.; Fang, Y.; Wu, X.; Liang, M.; Ding, X. Xenon Protects Against Septic Acute Kidney Injury via miR-21 Target Signaling Pathway. Crit. Care Med. 2015, 43, e250–e259. [Google Scholar] [PubMed]
  187. Xu, X.; Kriegel, A.J.; Liu, Y.; Usa, K.; Mladinov, D.; Liu, H.; Fang, Y.; Ding, X.; Liang, M. Delayed ischemic preconditioning contributes to renal protection by upregulation of miR-21. Kidney Int. 2012, 82, 1167–1175. [Google Scholar]
  188. Yang, G.; Pei, Y.; Cao, Q.; Wang, R. MicroRNA-21 represses human cystathionine gamma-lyase expression by targeting at specificity protein-1 in smooth muscle cells. J. Cell Physiol. 2012, 227, 3192–3200. [Google Scholar]
  189. Godwin, J.G.; Ge, X.; Stephan, K.; Jurisch, A.; Tullius, S.G.; Iacomini, J. Identification of a microRNA signature of renal ischemia reperfusion injury. Proc. Natl. Acad. Sci. USA 2010, 107, 14339–14344. [Google Scholar]
  190. Chen, C.; Lu, C.; Qian, Y.; Li, H.; Tan, Y.; Cai, L.; Weng, H. Urinary miR-21 as a potential biomarker of hypertensive kidney injury and fibrosis. Sci. Rep. 2017, 7, 17737. [Google Scholar]
  191. Kaucsar, T.; Revesz, C.; Godo, M.; Krenacs, T.; Albert, M.; Szalay, C.I.; Rosivall, L.; Benyo, Z.; Batkai, S.; Thum, T.; et al. Activation of the miR-17 family and miR-21 during murine kidney ischemia-reperfusion injury. Nucleic Acid. Ther. 2013, 23, 344–354. [Google Scholar]
  192. Saikumar, J.; Hoffmann, D.; Kim, T.M.; Gonzalez, V.R.; Zhang, Q.; Goering, P.L.; Brown, R.P.; Bijol, V.; Park, P.J.; Waikar, S.S.; et al. Expression, circulation, and excretion profile of microRNA-21, -155, and -18a following acute kidney injury. Toxicol. Sci. 2012, 129, 256–267. [Google Scholar] [PubMed]
  193. Pavkovic, M.; Riefke, B.; Ellinger-Ziegelbauer, H. Urinary microRNA profiling for identification of biomarkers after cisplatin-induced kidney injury. Toxicology 2014, 324, 147–157. [Google Scholar] [PubMed]
  194. Pu, X.Y.; Shen, J.Y.; Deng, Z.P.; Zhang, Z.A. Plasma-specific microRNA response induced by acute exposure to aristolochic acid I in rats. Arch. Toxicol. 2017, 91, 1473–1483. [Google Scholar]
  195. Ramachandran, K.; Saikumar, J.; Bijol, V.; Koyner, J.L.; Qian, J.; Betensky, R.A.; Waikar, S.S.; Vaidya, V.S. Human miRNome profiling identifies microRNAs differentially present in the urine after kidney injury. Clin. Chem. 2013, 59, 1742–1752. [Google Scholar]
  196. Du, J.; Cao, X.; Zou, L.; Chen, Y.; Guo, J.; Chen, Z.; Hu, S.; Zheng, Z. MicroRNA-21 and risk of severe acute kidney injury and poor outcomes after adult cardiac surgery. PLoS ONE 2013, 8, e63390. [Google Scholar]
  197. Gaede, L.; Liebetrau, C.; Blumenstein, J.; Troidl, C.; Dorr, O.; Kim, W.K.; Gottfried, K.; Voss, S.; Berkowitsch, A.; Walther, T.; et al. Plasma microRNA-21 for the early prediction of acute kidney injury in patients undergoing major cardiac surgery. Nephrol. Dial. Transpl. 2016, 31, 760–766. [Google Scholar]
  198. Wang, J.F.; Zha, Y.F.; Li, H.W.; Wang, F.; Bian, Q.; Lai, X.L.; Yu, G. Screening plasma miRNAs as biomarkers for renal ischemia-reperfusion injury in rats. Med. Sci. Monit. 2014, 20, 283–289. [Google Scholar]
  199. Wang, N.; Zhou, Y.; Jiang, L.; Li, D.; Yang, J.; Zhang, C.Y.; Zen, K. Urinary microRNA-10a and microRNA-30d serve as novel, sensitive and specific biomarkers for kidney injury. PLoS ONE 2012, 7, e51140. [Google Scholar]
  200. Ho, J.; Pandey, P.; Schatton, T.; Sims-Lucas, S.; Khalid, M.; Frank, M.H.; Hartwig, S.; Kreidberg, J.A. The pro-apoptotic protein Bim is a microRNA target in kidney progenitors. J. Am. Soc. Nephrol. 2011, 22, 1053–1063. [Google Scholar]
  201. Aguado-Fraile, E.; Ramos, E.; Conde, E.; Rodriguez, M.; Martin-Gomez, L.; Lietor, A.; Candela, A.; Ponte, B.; Liano, F.; Garcia-Bermejo, M.L. A Pilot Study Identifying a Set of microRNAs As Precise Diagnostic Biomarkers of Acute Kidney Injury. PLoS ONE 2015, 10, e0127175. [Google Scholar]
  202. Bhatt, K.; Zhou, L.; Mi, Q.S.; Huang, S.; She, J.X.; Dong, Z. MicroRNA-34a is induced via p53 during cisplatin nephrotoxicity and contributes to cell survival. Mol. Med. 2010, 16, 409–416. [Google Scholar] [PubMed]
  203. Finnerty, J.R.; Wang, W.X.; Hebert, S.S.; Wilfred, B.R.; Mao, G.; Nelson, P.T. The miR-15/107 group of microRNA genes: Evolutionary biology, cellular functions, and roles in human diseases. J. Mol. Biol. 2010, 402, 491–509. [Google Scholar]
  204. Wang, S.; Zhang, Z.; Wang, J.; Miao, H. MiR-107 induces TNF-alpha secretion in endothelial cells causing tubular cell injury in patients with septic acute kidney injury. Biochem. Biophys. Res. Commun. 2017, 483, 45–51. [Google Scholar] [PubMed]
  205. Weber, G.J.; Pushpakumar, S.B.; Sen, U. Hydrogen sulfide alleviates hypertensive kidney dysfunction through an epigenetic mechanism. Am. J. Physiol. Heart Circ. Physiol. 2017, 312, H874–H885. [Google Scholar]
  206. Lorenzen, J.M.; Kaucsar, T.; Schauerte, C.; Schmitt, R.; Rong, S.; Hubner, A.; Scherf, K.; Fiedler, J.; Martino, F.; Kumarswamy, R.; et al. MicroRNA-24 antagonism prevents renal ischemia reperfusion injury. J. Am. Soc. Nephrol. 2014, 25, 2717–2729. [Google Scholar] [PubMed]
  207. Kato, M.; Zhang, J.; Wang, M.; Lanting, L.; Yuan, H.; Rossi, J.J.; Natarajan, R. MicroRNA-192 in diabetic kidney glomeruli and its function in TGF-beta-induced collagen expression via inhibition of E-box repressors. Proc. Natl. Acad. Sci. USA 2007, 104, 3432–3437. [Google Scholar]
  208. Jung, M.; Mollenkopf, H.J.; Grimm, C.; Wagner, I.; Albrecht, M.; Waller, T.; Pilarsky, C.; Johannsen, M.; Stephan, C.; Lehrach, H.; et al. MicroRNA profiling of clear cell renal cell cancer identifies a robust signature to define renal malignancy. J. Cell Mol. Med. 2009, 13, 3918–3928. [Google Scholar]
  209. Zhu, H.Y.; Liu, M.Y.; Hong, Q.; Zhang, D.; Geng, W.J.; Xie, Y.S.; Chen, X.M. Role of microRNA-181a in the apoptosis of tubular epithelial cell induced by cisplatin. Chin. Med. J. 2012, 125, 523–526. [Google Scholar]
  210. Hou, D.; Wu, Q.; Wang, S.; Pang, S.; Liang, H.; Lyu, H.; Zhou, L.; Wang, Q.; Hao, L. Knockdown of miR-214 Alleviates Renal Interstitial Fibrosis by Targeting the Regulation of the PTEN/PI3K/AKT Signalling Pathway. Oxid. Med. Cell Longev. 2022, 2022, 7553928. [Google Scholar]
  211. Gluba-Sagr, A.; Franczyk, B.; Rysz-Gorzynska, M.; Lawinski, J.; Rysz, J. The Role of miRNA in Renal Fibrosis Leading to Chronic Kidney Disease. Biomedicines 2023, 11, 2358. [Google Scholar] [CrossRef]
  212. Denby, L.; Ramdas, V.; Lu, R.; Conway, B.R.; Grant, J.S.; Dickinson, B.; Aurora, A.B.; McClure, J.D.; Kipgen, D.; Delles, C.; et al. MicroRNA-214 antagonism protects against renal fibrosis. J. Am. Soc. Nephrol. 2014, 25, 65–80. [Google Scholar] [PubMed]
  213. Denby, L.; Ramdas, V.; McBride, M.W.; Wang, J.; Robinson, H.; McClure, J.; Crawford, W.; Lu, R.; Hillyard, D.Z.; Khanin, R.; et al. miR-21 and miR-214 are consistently modulated during renal injury in rodent models. Am. J. Pathol. 2011, 179, 661–672. [Google Scholar] [PubMed]
  214. Cui, R.; Xu, J.; Chen, X.; Zhu, W. Global miRNA expression is temporally correlated with acute kidney injury in mice. PeerJ 2016, 4, e1729. [Google Scholar] [PubMed]
  215. Lan, Y.F.; Chen, H.H.; Lai, P.F.; Cheng, C.F.; Huang, Y.T.; Lee, Y.C.; Chen, T.W.; Lin, H. MicroRNA-494 reduces ATF3 expression and promotes AKI. J. Am. Soc. Nephrol. 2012, 23, 2012–2023. [Google Scholar] [PubMed]
  216. Bhatt, K.; Wei, Q.; Pabla, N.; Dong, G.; Mi, Q.S.; Liang, M.; Mei, C.; Dong, Z. MicroRNA-687 Induced by Hypoxia-Inducible Factor-1 Targets Phosphatase and Tensin Homolog in Renal Ischemia-Reperfusion Injury. J. Am. Soc. Nephrol. 2015, 26, 1588–1596. [Google Scholar]
  217. Ge, Q.M.; Huang, C.M.; Zhu, X.Y.; Bian, F.; Pan, S.M. Differentially expressed miRNAs in sepsis-induced acute kidney injury target oxidative stress and mitochondrial dysfunction pathways. PLoS ONE 2017, 12, e0173292. [Google Scholar]
  218. Fan, P.C.; Chen, C.C.; Peng, C.C.; Chang, C.H.; Yang, C.H.; Yang, C.; Chu, L.J.; Chen, Y.C.; Yang, C.W.; Chang, Y.S.; et al. A circulating miRNA signature for early diagnosis of acute kidney injury following acute myocardial infarction. J. Transl. Med. 2019, 17, 139. [Google Scholar]
  219. Gutierrez-Escolano, A.; Santacruz-Vazquez, E.; Gomez-Perez, F. Dysregulated microRNAs involved in contrast-induced acute kidney injury in rat and human. Ren. Fail. 2015, 37, 1498–1506. [Google Scholar]
  220. Sun, S.Q.; Zhang, T.; Ding, D.; Zhang, W.F.; Wang, X.L.; Sun, Z.; Hu, L.H.; Qin, S.Y.; Shen, L.H.; He, B. Circulating MicroRNA-188, -30a, and -30e as Early Biomarkers for Contrast-Induced Acute Kidney Injury. J. Am. Heart Assoc. 2016, 5. [Google Scholar]
  221. Zou, Y.F.; Wen, D.; Zhao, Q.; Shen, P.Y.; Shi, H.; Zhao, Q.; Chen, Y.X.; Zhang, W. Urinary MicroRNA-30c-5p and MicroRNA-192-5p as potential biomarkers of ischemia-reperfusion-induced kidney injury. Exp. Biol. Med. 2017, 242, 657–667. [Google Scholar]
  222. Lorenzen, J.M.; Kielstein, J.T.; Hafer, C.; Gupta, S.K.; Kumpers, P.; Faulhaber-Walter, R.; Haller, H.; Fliser, D.; Thum, T. Circulating miR-210 predicts survival in critically ill patients with acute kidney injury. Clin. J. Am. Soc. Nephrol. 2011, 6, 1540–1546. [Google Scholar] [PubMed]
  223. Zhou, X.; Qu, Z.; Zhu, C.; Lin, Z.; Huo, Y.; Wang, X.; Wang, J.; Li, B. Identification of urinary microRNA biomarkers for detection of gentamicin-induced acute kidney injury in rats. Regul. Toxicol. Pharmacol. 2016, 78, 78–84. [Google Scholar]
  224. Wei, Q.; Sun, H.; Song, S.; Liu, Y.; Liu, P.; Livingston, M.J.; Wang, J.; Liang, M.; Mi, Q.S.; Huo, Y.; et al. MicroRNA-668 represses MTP18 to preserve mitochondrial dynamics in ischemic acute kidney injury. J. Clin. Investig. 2018, 128, 5448–5464. [Google Scholar]
  225. Wallace, J.L. Physiological and pathophysiological roles of hydrogen sulfide in the gastrointestinal tract. Antioxid. Redox Signal 2010, 12, 1125–1133. [Google Scholar] [PubMed]
  226. Wang, R. Hydrogen sulfide: A new EDRF. Kidney Int. 2009, 76, 700–704. [Google Scholar] [PubMed]
  227. Whiteman, M.; Winyard, P.G. Hydrogen sulfide and inflammation: The good, the bad, the ugly and the promising. Expert. Rev. Clin. Pharmacol. 2011, 4, 13–32. [Google Scholar]
  228. Wallace, J.L.; Vong, L.; McKnight, W.; Dicay, M.; Martin, G.R. Endogenous and exogenous hydrogen sulfide promotes resolution of colitis in rats. Gastroenterology 2009, 137, 569–578.e1. [Google Scholar]
  229. Wallace, J.L.; Dicay, M.; McKnight, W.; Martin, G.R. Hydrogen sulfide enhances ulcer healing in rats. FASEB J. 2007, 21, 4070–4076. [Google Scholar]
  230. Wang, X.; Lin, T.; He, Y.; Zhou, Y.; Peng, Y.; Zhang, W.; Ni, X. Reduced Renal CSE/CBS/H2S Contributes to the Progress of Lupus Nephritis. Biology 2023, 12, 318. [Google Scholar] [CrossRef]
  231. Zhang, L.; Rao, J.; Liu, X.; Wang, X.; Wang, C.; Fu, S.; Xiao, J. Attenuation of Sepsis-Induced Acute Kidney Injury by Exogenous H(2)S via Inhibition of Ferroptosis. Molecules 2023, 28, 4770. [Google Scholar] [CrossRef]
  232. Pushpakumar, S.; Kundu, S.; Weber, G.; Sen, U. Exogenous hydrogen sulfide and miR-21 antagonism attenuates macrophage-mediated inflammation in ischemia reperfusion injury of the aged kidney. GeroScience 2021, 43, 1349–1367. [Google Scholar] [PubMed]
  233. Teksen, Y.; Kadioglu, E.; Kocak, C.; Kocak, H. Effect of Hydrogen Sulfide on Kidney Injury in Rat Model of Crush Syndrome. J. Surg. Res. 2019, 235, 470–478. [Google Scholar] [PubMed]
  234. Zhi, L.; Ang, A.D.; Zhang, H.; Moore, P.K.; Bhatia, M. Hydrogen sulfide induces the synthesis of proinflammatory cytokines in human monocyte cell line U937 via the ERK-NF-kappaB pathway. J. Leukoc. Biol. 2007, 81, 1322–1332. [Google Scholar]
  235. Zhu, X.Y.; Liu, S.J.; Liu, Y.J.; Wang, S.; Ni, X. Glucocorticoids suppress cystathionine gamma-lyase expression and H2S production in lipopolysaccharide-treated macrophages. Cell Mol. Life Sci. 2010, 67, 1119–1132. [Google Scholar] [PubMed]
  236. Lee, M.; Sparatore, A.; Del Soldato, P.; McGeer, E.; McGeer, P.L. Hydrogen sulfide-releasing NSAIDs attenuate neuroinflammation induced by microglial and astrocytic activation. Glia 2010, 58, 103–113. [Google Scholar]
  237. Maclean, K.N.; Sikora, J.; Kozich, V.; Jiang, H.; Greiner, L.S.; Kraus, E.; Krijt, J.; Crnic, L.S.; Allen, R.H.; Stabler, S.P.; et al. Cystathionine beta-synthase null homocystinuric mice fail to exhibit altered hemostasis or lowering of plasma homocysteine in response to betaine treatment. Mol. Genet. Metab. 2010, 101, 163–171. [Google Scholar]
  238. Ishii, I.; Akahoshi, N.; Yamada, H.; Nakano, S.; Izumi, T.; Suematsu, M. Cystathionine gamma-Lyase-deficient mice require dietary cysteine to protect against acute lethal myopathy and oxidative injury. J. Biol. Chem. 2010, 285, 26358–26368. [Google Scholar]
  239. Lameire, N.H.; Bagga, A.; Cruz, D.; De Maeseneer, J.; Endre, Z.; Kellum, J.A.; Liu, K.D.; Mehta, R.L.; Pannu, N.; Van Biesen, W.; et al. Acute kidney injury: An increasing global concern. Lancet 2013, 382, 170–179. [Google Scholar]
  240. Bonventre, J.V.; Yang, L. Cellular pathophysiology of ischemic acute kidney injury. J. Clin. Investig. 2011, 121, 4210–4221. [Google Scholar]
  241. Eltzschig, H.K.; Eckle, T. Ischemia and reperfusion--from mechanism to translation. Nat. Med. 2011, 17, 1391–1401. [Google Scholar]
  242. Kalogeris, T.; Baines, C.P.; Krenz, M.; Korthuis, R.J. Cell biology of ischemia/reperfusion injury. Int. Rev. Cell Mol. Biol. 2012, 298, 229–317. [Google Scholar] [PubMed]
  243. Thurman, J.M. Triggers of inflammation after renal ischemia/reperfusion. Clin. Immunol. 2007, 123, 7–13. [Google Scholar] [PubMed]
  244. Mills, C.D. M1 and M2 Macrophages: Oracles of Health and Disease. Crit. Rev. Immunol. 2012, 32, 463–488. [Google Scholar] [PubMed]
  245. Dufton, N.; Natividad, J.; Verdu, E.F.; Wallace, J.L. Hydrogen sulfide and resolution of acute inflammation: A comparative study utilizing a novel fluorescent probe. Sci. Rep. 2012, 2, 499. [Google Scholar]
  246. Whiteman, M.; Li, L.; Rose, P.; Tan, C.H.; Parkinson, D.B.; Moore, P.K. The effect of hydrogen sulfide donors on lipopolysaccharide-induced formation of inflammatory mediators in macrophages. Antioxid. Redox Signal. 2010, 12, 1147–1154. [Google Scholar] [PubMed]
  247. Nelson, P.J.; Rees, A.J.; Griffin, M.D.; Hughes, J.; Kurts, C.; Duffield, J. The renal mononuclear phagocytic system. J. Am. Soc. Nephrol. 2012, 23, 194–203. [Google Scholar]
  248. Dong, X.; Swaminathan, S.; Bachman, L.A.; Croatt, A.J.; Nath, K.A.; Griffin, M.D. Resident dendritic cells are the predominant TNF-secreting cell in early renal ischemia-reperfusion injury. Kidney Int. 2007, 71, 619–628. [Google Scholar]
  249. Li, L.; Huang, L.; Sung, S.S.; Vergis, A.L.; Rosin, D.L.; Rose, C.E.; Jr Lobo, P.I.; Okusa, M.D. The chemokine receptors CCR2 and CX3CR1 mediate monocyte/macrophage trafficking in kidney ischemia-reperfusion injury. Kidney Int. 2008, 74, 1526–1537. [Google Scholar]
  250. Tittel, A.P.; Heuser, C.; Ohliger, C.; Knolle, P.A.; Engel, D.R.; Kurts, C. Kidney dendritic cells induce innate immunity against bacterial pyelonephritis. J. Am. Soc. Nephrol. 2011, 22, 1435–1441. [Google Scholar]
  251. Serbina, N.V.; Pamer, E.G. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat. Immunol. 2006, 7, 311–317. [Google Scholar]
  252. Cao, Q.; Harris, D.C.; Wang, Y. Macrophages in kidney injury, inflammation, and fibrosis. Physiology 2015, 30, 183–194. [Google Scholar] [PubMed]
  253. Lee, S.; Huen, S.; Nishio, H.; Nishio, S.; Lee, H.K.; Choi, B.S.; Ruhrberg, C.; Cantley, L.G. Distinct macrophage phenotypes contribute to kidney injury and repair. J. Am. Soc. Nephrol. 2011, 22, 317–326. [Google Scholar] [PubMed]
  254. Kurts, C.; Panzer, U.; Anders, H.J.; Rees, A.J. The immune system and kidney disease: Basic concepts and clinical implications. Nat. Rev. Immunol. 2013, 13, 738–753. [Google Scholar]
  255. Jang, H.R.; Rabb, H. The innate immune response in ischemic acute kidney injury. Clin. Immunol. 2009, 130, 41–50. [Google Scholar]
  256. Lu, G.; Zhang, R.; Geng, S.; Peng, L.; Jayaraman, P.; Chen, C.; Xu, F.; Yang, J.; Li, Q.; Zheng, H.; et al. Myeloid cell-derived inducible nitric oxide synthase suppresses M1 macrophage polarization. Nat. Commun. 2015, 6, 6676. [Google Scholar]
  257. Muraille, E.; Leo, O.; Moser, M. TH1/TH2 paradigm extended: Macrophage polarization as an unappreciated pathogen-driven escape mechanism? Front. Immunol. 2014, 5, 603. [Google Scholar]
  258. Cho, W.Y.; Choi, H.M.; Lee, S.Y.; Kim, M.G.; Kim, H.K.; Jo, S.K. The role of Tregs and CD11c(+) macrophages/dendritic cells in ischemic preconditioning of the kidney. Kidney Int. 2010, 78, 981–992. [Google Scholar]
  259. Kim, M.G.; Boo, C.S.; Ko, Y.S.; Lee, H.Y.; Cho, W.Y.; Kim, H.K.; Jo, S.K. Depletion of kidney CD11c+ F4/80+ cells impairs the recovery process in ischaemia/reperfusion-induced acute kidney injury. Nephrol. Dial. Transpl. 2010, 25, 2908–2921. [Google Scholar]
  260. Weisheit, C.K.; Engel, D.R.; Kurts, C. Dendritic Cells and Macrophages: Sentinels in the Kidney. Clin. J. Am. Soc. Nephrol. 2015, 10, 1841–1851. [Google Scholar]
  261. Geissmann, F.; Jung, S.; Littman, D.R. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 2003, 19, 71–82. [Google Scholar]
  262. Guilliams, M.; Mildner, A.; Yona, S. Developmental and Functional Heterogeneity of Monocytes. Immunity 2018, 49, 595–613. [Google Scholar] [PubMed]
  263. Thomas, G.; Tacke, R.; Hedrick, C.C.; Hanna, R.N. Nonclassical patrolling monocyte function in the vasculature. Arter. Thromb. Vasc. Biol. 2015, 35, 1306–1316. [Google Scholar]
  264. Bystrom, J.; Evans, I.; Newson, J.; Stables, M.; Toor, I.; van Rooijen, N.; Crawford, M.; Colville-Nash, P.; Farrow, S.; Gilroy, D.W. Resolution-phase macrophages possess a unique inflammatory phenotype that is controlled by cAMP. Blood 2008, 112, 4117–4127. [Google Scholar]
  265. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [PubMed]
  266. Kinsey, G.R.; Sharma, R.; Huang, L.; Li, L.; Vergis, A.L.; Ye, H.; Ju, S.T.; Okusa, M.D. Regulatory T cells suppress innate immunity in kidney ischemia-reperfusion injury. J. Am. Soc. Nephrol. 2009, 20, 1744–1753. [Google Scholar]
  267. Jang, H.R.; Rabb, H. Immune cells in experimental acute kidney injury. Nat. Rev. Nephrol. 2015, 11, 88–101. [Google Scholar]
  268. Sharfuddin, A.A.; Molitoris, B.A. Pathophysiology of ischemic acute kidney injury. Nat. Rev. Nephrol. 2011, 7, 189–200. [Google Scholar]
  269. Wegiel, B.; Hedblom, A.; Li, M.; Gallo, D.; Csizmadia, E.; Harris, C.; Nemeth, Z.; Zuckerbraun, B.S.; Soares, M.; Persson, J.L.; et al. Heme oxygenase-1 derived carbon monoxide permits maturation of myeloid cells. Cell Death Dis. 2014, 5, e1139. [Google Scholar]
  270. Anders, H.J.; Ryu, M. Renal microenvironments and macrophage phenotypes determine progression or resolution of renal inflammation and fibrosis. Kidney Int. 2011, 80, 915–925. [Google Scholar]
  271. Galli, S.J.; Borregaard, N.; Wynn, T.A. Phenotypic and functional plasticity of cells of innate immunity: Macrophages, mast cells and neutrophils. Nat. Immunol. 2011, 12, 1035–1044. [Google Scholar]
  272. Duffield, J.S. Macrophages and immunologic inflammation of the kidney. Semin. Nephrol. 2010, 30, 234–254. [Google Scholar] [PubMed]
  273. Murray, P.J.; Wynn, T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011, 11, 723–737. [Google Scholar] [PubMed]
  274. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar]
  275. Clements, M.; Gershenovich, M.; Chaber, C.; Campos-Rivera, J.; Du, P.; Zhang, M.; Ledbetter, S.; Zuk, A. Differential Ly6C Expression after Renal Ischemia-Reperfusion Identifies Unique Macrophage Populations. J. Am. Soc. Nephrol. 2016, 27, 159–170. [Google Scholar]
  276. Huet, E.; Gabison, E.E.; Mourah, S.; Menashi, S. Role of emmprin/CD147 in tissue remodeling. Connect. Tissue Res. 2008, 49, 175–179. [Google Scholar]
  277. Gabison, E.E.; Hoang-Xuan, T.; Mauviel, A.; Menashi, S. EMMPRIN/CD147, an MMP modulator in cancer, development and tissue repair. Biochimie 2005, 87, 361–368. [Google Scholar] [PubMed]
  278. Cabral-Pacheco, G.A.; Garza-Veloz, I.; Castruita-De la Rosa, C.; Ramirez-Acuna, J.M.; Perez-Romero, B.A.; Guerrero-Rodriguez, J.F.; Martinez-Avila, N.; Martinez-Fierro, M.L. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int. J. Mol. Sci. 2020, 21, 9739. [Google Scholar] [CrossRef]
  279. Tarin, C.; Lavin, B.; Gomez, M.; Saura, M.; Diez-Juan, A.; Zaragoza, C. The extracellular matrix metalloproteinase inducer EMMPRIN is a target of nitric oxide in myocardial ischemia/reperfusion. Free Radic. Biol. Med. 2011, 51, 387–395. [Google Scholar]
  280. Beynon, R.J.; Shannon, J.D.; Bond, J.S. Purification and characterization of a metallo-endoproteinase from mouse kidney. Biochem. J. 1981, 199, 591–598. [Google Scholar]
  281. Kenny, A.J.; Ingram, J. Proteins of the kidney microvillar membrane. Purification and properties of the phosphoramidon-insensitive endopeptidase (‘endopeptidase-2’) from rat kidney. Biochem. J. 1987, 245, 515–524. [Google Scholar]
  282. Sterchi, E.E.; Green, J.R.; Lentze, M.J. Non-pancreatic hydrolysis of N-benzoyl-l-tyrosyl-p-aminobenzoic acid (PABA-peptide) in the human small intestine. Clin. Sci. 1982, 62, 557–560. [Google Scholar]
  283. Geurts, N.; Becker-Pauly, C.; Martens, E.; Proost, P.; Van den Steen, P.E.; Stocker, W.; Opdenakker, G. Meprins process matrix metalloproteinase-9 (MMP-9)/gelatinase B and enhance the activation kinetics by MMP-3. FEBS Lett. 2012, 586, 4264–4269. [Google Scholar] [PubMed]
  284. Kaushal, G.P.; Haun, R.S.; Herzog, C.; Shah, S.V. Meprin A metalloproteinase and its role in acute kidney injury. Am. J. Physiol. Ren. Physiol. 2013, 304, F1150–F1158. [Google Scholar]
  285. Walker, P.D.; Kaushal, G.P.; Shah, S.V. Meprin A, the major matrix degrading enzyme in renal tubules, produces a novel nidogen fragment in vitro and in vivo. Kidney Int. 1998, 53, 1673–1680. [Google Scholar]
  286. Bylander, J.; Li, Q.; Ramesh, G.; Zhang, B.; Reeves, W.B.; Bond, J.S. Targeted disruption of the meprin metalloproteinase beta gene protects against renal ischemia-reperfusion injury in mice. Am. J. Physiol. Ren. Physiol. 2008, 294, F480–F490. [Google Scholar]
  287. Basile, D.P.; Fredrich, K.; Weihrauch, D.; Hattan, N.; Chilian, W.M. Angiostatin and matrix metalloprotease expression following ischemic acute renal failure. Am. J. Physiol. Ren. Physiol. 2004, 286, F893–F902. [Google Scholar]
  288. Caron, A.; Desrosiers, R.R.; Langlois, S.; Beliveau, R. Ischemia-reperfusion injury stimulates gelatinase expression and activity in kidney glomeruli. Can. J. Physiol. Pharmacol. 2005, 83, 287–300. [Google Scholar] [PubMed]
  289. Kunugi, S.; Shimizu, A.; Kuwahara, N.; Du, X.; Takahashi, M.; Terasaki, Y.; Fujita, E.; Mii, A.; Nagasaka, S.; Akimoto, T.; et al. Inhibition of matrix metalloproteinases reduces ischemia-reperfusion acute kidney injury. Lab. Investig. 2011, 91, 170–180. [Google Scholar]
  290. Winchester, L.J.; Veeranki, S.; Givvimani, S.; Tyagi, S.C. Homocysteine elicits an M1 phenotype in murine macrophages through an EMMPRIN-mediated pathway. Can. J. Physiol. Pharmacol. 2015, 93, 577–584. [Google Scholar]
  291. Huen, S.C.; Cantley, L.G. Macrophages in Renal Injury and Repair. Annu. Rev. Physiol. 2017, 79, 449–469. [Google Scholar]
  292. Toba, H.; Lindsey, M.L. Extracellular matrix roles in cardiorenal fibrosis: Potential therapeutic targets for CVD and CKD in the elderly. Pharmacol. Ther. 2019, 193, 99–120. [Google Scholar] [PubMed]
  293. Wang, Y.; Xing, Q.Q.; Tu, J.K.; Tang, W.B.; Yuan, X.N.; Xie, Y.Y.; Wang, W.; Peng, Z.Z.; Huang, L.; Xu, H.; et al. Involvement of hydrogen sulfide in the progression of renal fibrosis. Chin. Med. J. 2019, 132, 2872–2880. [Google Scholar] [PubMed]
  294. Pushpakumar, S.; Kundu, S.; Narayanan, N.; Sen, U. DNA hypermethylation in hyperhomocysteinemia contributes to abnormal extracellular matrix metabolism in the kidney. FASEB J. 2015, 29, 4713–4725. [Google Scholar] [PubMed]
  295. Hayashi, T.; Takai, S.; Yamashita, C. Impact of the renin-angiotensin-aldosterone-system on cardiovascular and renal complications in diabetes mellitus. Curr. Vasc. Pharmacol. 2010, 8, 189–197. [Google Scholar]
  296. Zeng, O.; Li, F.; Li, Y.; Li, L.; Xiao, T.; Chu, C.; Yang, J. Effect of Novel Gasotransmitter hydrogen sulfide on renal fibrosis and connexins expression in diabetic rats. Bioengineered 2016, 7, 314–320. [Google Scholar]
  297. Kundu, S.; Pushpakumar, S.; Sen, U. MMP-9- and NMDA receptor-mediated mechanism of diabetic renovascular remodeling and kidney dysfunction: Hydrogen sulfide is a key modulator. Nitric Oxide 2015, 46, 172–185. [Google Scholar]
  298. Kundu, S.; Pushpakumar, S.B.; Tyagi, A.; Coley, D.; Sen, U. Hydrogen sulfide deficiency and diabetic renal remodeling: Role of matrix metalloproteinase-9. Am. J. Physiol. Endocrinol. Metab. 2013, 304, E1365–E1378. [Google Scholar]
  299. Ngowi, E.E.; Sarfraz, M.; Afzal, A.; Khan, N.H.; Khattak, S.; Zhang, X.; Li, T.; Duan, S.F.; Ji, X.Y.; Wu, D.D. Roles of Hydrogen Sulfide Donors in Common Kidney Diseases. Front. Pharmacol. 2020, 11, 564281. [Google Scholar]
  300. Lenz, O.; Elliot, S.J.; Stetler-Stevenson, W.G. Matrix metalloproteinases in renal development and disease. J. Am. Soc. Nephrol. 2000, 11, 574–581. [Google Scholar]
  301. Ahmed, A.K.; Haylor, J.L.; El Nahas, A.M.; Johnson, T.S. Localization of matrix metalloproteinases and their inhibitors in experimental progressive kidney scarring. Kidney Int. 2007, 71, 755–763. [Google Scholar]
  302. Border, W.A.; Noble, N.A. TGF-beta in kidney fibrosis: A target for gene therapy. Kidney Int. 1997, 51, 1388–1396. [Google Scholar] [PubMed]
  303. Werb, Z.; Hembry, R.M.; Murphy, G.; Aggeler, J. Commitment to expression of the metalloendopeptidases, collagenase and stromelysin: Relationship of inducing events to changes in cytoskeletal architecture. J. Cell Biol. 1986, 102, 697–702. [Google Scholar] [PubMed]
  304. Surendran, K.; Simon, T.C.; Liapis, H.; McGuire, J.K. Matrilysin (MMP-7) expression in renal tubular damage: Association with Wnt4. Kidney Int. 2004, 65, 2212–2222. [Google Scholar] [PubMed]
  305. Uchio, K.; Sawada, K.; Manabe, N. Expression of macrophage metalloelastase (MMP-12) in podocytes of hereditary nephrotic mice (ICGN strain). J. Vet. Med. Sci. 2009, 71, 305–312. [Google Scholar] [PubMed]
  306. Yang, Z.Z.; Zou, A.P. Homocysteine enhances TIMP-1 expression and cell proliferation associated with NADH oxidase in rat mesangial cells. Kidney Int. 2003, 63, 1012–1020. [Google Scholar]
  307. Sharma, R.; Suzuki, K.; Nagase, H.; Savin, V.J. Matrix metalloproteinase (stromelysin-1) increases the albumin permeability of isolated rat glomeruli. J. Lab. Clin. Med. 1996, 128, 297–303. [Google Scholar]
  308. Torres, L.; Garcia-Trevijano, E.R.; Rodriguez, J.A.; Carretero, M.V.; Bustos, M.; Fernandez, E.; Eguinoa, E.; Mato, J.M.; Avila, M.A. Induction of TIMP-1 expression in rat hepatic stellate cells and hepatocytes: A new role for homocysteine in liver fibrosis. Biochim. Biophys. Acta 1999, 1455, 12–22. [Google Scholar]
  309. Altaany, Z.; Moccia, F.; Munaron, L.; Mancardi, D.; Wang, R. Hydrogen sulfide and endothelial dysfunction: Relationship with nitric oxide. Curr. Med. Chem. 2014, 21, 3646–3661. [Google Scholar]
  310. Fiorucci, S.; Antonelli, E.; Distrutti, E.; Rizzo, G.; Mencarelli, A.; Orlandi, S.; Zanardo, R.; Renga, B.; Di Sante, M.; Morelli, A.; et al. Inhibition of hydrogen sulfide generation contributes to gastric injury caused by anti-inflammatory nonsteroidal drugs. Gastroenterology 2005, 129, 1210–1224. [Google Scholar]
  311. Zuidema, M.Y.; Yang, Y.; Wang, M.; Kalogeris, T.; Liu, Y.; Meininger, C.J.; Hill, M.A.; Davis, M.J.; Korthuis, R.J. Antecedent hydrogen sulfide elicits an anti-inflammatory phenotype in postischemic murine small intestine: Role of BK channels. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H1554–H1567. [Google Scholar]
  312. Muzaffar, S.; Jeremy, J.Y.; Sparatore, A.; Del Soldato, P.; Angelini, G.D.; Shukla, N. H2S-donating sildenafil (ACS6) inhibits superoxide formation and gp91phox expression in arterial endothelial cells: Role of protein kinases A and G. Br. J. Pharmacol. 2008, 155, 984–994. [Google Scholar] [PubMed]
  313. Suo, R.; Zhao, Z.Z.; Tang, Z.H.; Ren, Z.; Liu, X.; Liu, L.S.; Wang, Z.; Tang, C.K.; Wei, D.H.; Jiang, Z.S. Hydrogen sulfide prevents H(2)O(2)-induced senescence in human umbilical vein endothelial cells through SIRT1 activation. Mol. Med. Rep. 2013, 7, 1865–1870. [Google Scholar] [PubMed]
  314. Yang, G.; Zhao, K.; Ju, Y.; Mani, S.; Cao, Q.; Puukila, S.; Khaper, N.; Wu, L.; Wang, R. Hydrogen sulfide protects against cellular senescence via S-sulfhydration of Keap1 and activation of Nrf2. Antioxid. Redox. Signal. 2013, 18, 1906–1919. [Google Scholar] [PubMed]
  315. Yan, S.K.; Chang, T.; Wang, H.; Wu, L.; Wang, R.; Meng, Q.H. Effects of hydrogen sulfide on homocysteine-induced oxidative stress in vascular smooth muscle cells. Biochem. Biophys. Res. Commun. 2006, 351, 485–491. [Google Scholar] [PubMed]
  316. Carden, D.L.; Granger, D.N. Pathophysiology of ischaemia-reperfusion injury. J. Pathol. 2000, 190, 255–266. [Google Scholar]
  317. Eltzschig, H.K.; Collard, C.D. Vascular ischaemia and reperfusion injury. Br. Med. Bull. 2004, 70, 71–86. [Google Scholar]
  318. Streeter, E.; Hart, J.; Badoer, E. An investigation of the mechanisms of hydrogen sulfide-induced vasorelaxation in rat middle cerebral arteries. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2012, 385, 991–1002. [Google Scholar]
  319. Bibli, S.I.; Hu, J.; Sigala, F.; Wittig, I.; Heidler, J.; Zukunft, S.; Tsilimigras, D.I.; Randriamboavonjy, V.; Wittig, J.; Kojonazarov, B.; et al. Cystathionine gamma Lyase Sulfhydrates the RNA Binding Protein Human Antigen R to Preserve Endothelial Cell Function and Delay Atherogenesis. Circulation 2019, 139, 101–114. [Google Scholar]
  320. Coletta, C.; Modis, K.; Szczesny, B.; Brunyanszki, A.; Olah, G.; Rios, E.C.; Yanagi, K.; Ahmad, A.; Papapetropoulos, A.; Szabo, C. Regulation of Vascular Tone, Angiogenesis and Cellular Bioenergetics by the 3-Mercaptopyruvate Sulfurtransferase/H2S Pathway: Functional Impairment by Hyperglycemia and Restoration by DL-alpha-Lipoic Acid. Mol. Med. 2015, 21, 1–14. [Google Scholar]
  321. Ciccone, V.; Genah, S.; Morbidelli, L. Endothelium as a Source and Target of H(2)S to Improve Its Trophism and Function. Antioxidants 2021, 10, 486. [Google Scholar] [CrossRef]
  322. Kovacic, J.C.; Dimmeler, S.; Harvey, R.P.; Finkel, T.; Aikawa, E.; Krenning, G.; Baker, A.H. Endothelial to Mesenchymal Transition in Cardiovascular Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 190–209. [Google Scholar] [PubMed]
  323. Piera-Velazquez, S.; Li, Z.; Jimenez, S.A. Role of endothelial-mesenchymal transition (EndoMT) in the pathogenesis of fibrotic disorders. Am. J. Pathol. 2011, 179, 1074–1080. [Google Scholar] [PubMed]
  324. Rieder, F.; Kessler, S.P.; West, G.A.; Bhilocha, S.; de la Motte, C.; Sadler, T.M.; Gopalan, B.; Stylianou, E.; Fiocchi, C. Inflammation-induced endothelial-to-mesenchymal transition: A novel mechanism of intestinal fibrosis. Am. J. Pathol. 2011, 179, 2660–2673. [Google Scholar]
  325. Zeisberg, E.M.; Tarnavski, O.; Zeisberg, M.; Dorfman, A.L.; McMullen, J.R.; Gustafsson, E.; Chandraker, A.; Yuan, X.; Pu, W.T.; Roberts, A.B.; et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat. Med. 2007, 13, 952–961. [Google Scholar] [PubMed]
  326. Kizu, A.; Medici, D.; Kalluri, R. Endothelial-mesenchymal transition as a novel mechanism for generating myofibroblasts during diabetic nephropathy. Am. J. Pathol. 2009, 175, 1371–1373. [Google Scholar] [PubMed]
  327. Zeisberg, E.M.; Potenta, S.E.; Sugimoto, H.; Zeisberg, M.; Kalluri, R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J. Am. Soc. Nephrol. 2008, 19, 2282–2287. [Google Scholar]
  328. Jinnin, M.; Ihn, H.; Tamaki, K. Characterization of SIS3, a novel specific inhibitor of Smad3, and its effect on transforming growth factor-β1-induced extracellular matrix expression. Mol. Pharmacol. 2006, 69, 597–607. [Google Scholar]
  329. Genovese, F.; Manresa, A.A.; Leeming, D.J.; Karsdal, M.A.; Boor, P. The extracellular matrix in the kidney: A source of novel non-invasive biomarkers of kidney fibrosis? Fibrogenesis Tissue Repair. 2014, 7, 4. [Google Scholar]
  330. Johnson, T.S.; Haylor, J.L.; Thomas, G.L.; Fisher, M.; El Nahas, A.M. Matrix metalloproteinases and their inhibitions in experimental renal scarring. Exp. Nephrol. 2002, 10, 182–195. [Google Scholar]
  331. Sharma, A.K.; Mauer, S.M.; Kim, Y.; Michael, A.F. Interstitial fibrosis in obstructive nephropathy. Kidney Int. 1993, 44, 774–788. [Google Scholar]
  332. Bakun, M.; Niemczyk, M.; Domanski, D.; Jazwiec, R.; Perzanowska, A.; Niemczyk, S.; Kistowski, M.; Fabijanska, A.; Borowiec, A.; Paczek, L.; et al. Urine proteome of autosomal dominant polycystic kidney disease patients. Clin. Proteom. 2012, 9, 13. [Google Scholar]
  333. Patschan, D.; Schwarze, K.; Henze, E.; Patschan, S.; Muller, G.A. Endothelial autophagy and Endothelial-to-Mesenchymal Transition (EndoMT) in eEPC treatment of ischemic AKI. J. Nephrol. 2016, 29, 637–644. [Google Scholar] [PubMed]
  334. Khwaja, A. KDIGO clinical practice guidelines for acute kidney injury. Nephron Clin. Pract. 2012, 120, c179–c184. [Google Scholar] [PubMed]
  335. Pickkers, P.; Ostermann, M.; Joannidis, M.; Zarbock, A.; Hoste, E.; Bellomo, R.; Prowle, J.; Darmon, M.; Bonventre, J.V.; Forni, L.; et al. The intensive care medicine agenda on acute kidney injury. Intensive Care Med. 2017, 43, 1198–1209. [Google Scholar] [PubMed]
  336. Moore, P.K.; Hsu, R.K.; Liu, K.D. Management of Acute Kidney Injury: Core Curriculum 2018. Am. J. Kidney Dis. 2018, 72, 136–148. [Google Scholar] [PubMed]
  337. Ostermann, M.; Liu, K.; Kashani, K. Fluid Management in Acute Kidney Injury. Chest 2019, 156, 594–603. [Google Scholar]
  338. Meersch, M.; Schmidt, C.; Hoffmeier, A.; Van Aken, H.; Wempe, C.; Gerss, J.; Zarbock, A. Prevention of cardiac surgery-associated AKI by implementing the KDIGO guidelines in high risk patients identified by biomarkers: The PrevAKI randomized controlled trial. Intensive Care Med. 2017, 43, 1551–1561. [Google Scholar]
  339. Bagshaw, S.M.; Lapinsky, S.; Dial, S.; Arabi, Y.; Dodek, P.; Wood, G.; Ellis, P.; Guzman, J.; Marshall, J.; Parrillo, J.E.; et al. Acute kidney injury in septic shock: Clinical outcomes and impact of duration of hypotension prior to initiation of antimicrobial therapy. Intensive Care Med. 2009, 35, 871–881. [Google Scholar]
  340. Varrier, M.; Ostermann, M. Fluid Composition and Clinical Effects. Crit. Care Clin. 2015, 31, 823–837. [Google Scholar]
  341. Myburgh, J.A.; Finfer, S.; Bellomo, R.; Billot, L.; Cass, A.; Gattas, D.; Glass, P.; Lipman, J.; Liu, B.; McArthur, C.; et al. Hydroxyethyl starch or saline for fluid resuscitation in intensive care. N. Engl. J. Med. 2012, 367, 1901–1911. [Google Scholar]
  342. Asfar, P.; Meziani, F.; Hamel, J.F.; Grelon, F.; Megarbane, B.; Anguel, N.; Mira, J.P.; Dequin, P.F.; Gergaud, S.; Weiss, N.; et al. High versus low blood-pressure target in patients with septic shock. N. Engl. J. Med. 2014, 370, 1583–1593. [Google Scholar] [PubMed]
  343. Kato, R.; Pinsky, M.R. Personalizing blood pressure management in septic shock. Ann. Intensive Care 2015, 5, 41. [Google Scholar] [PubMed]
  344. Futier, E.; Lefrant, J.Y.; Guinot, P.G.; Godet, T.; Lorne, E.; Cuvillon, P.; Bertran, S.; Leone, M.; Pastene, B.; Piriou, V.; et al. Effect of Individualized vs Standard Blood Pressure Management Strategies on Postoperative Organ Dysfunction Among High-Risk Patients Undergoing Major Surgery: A Randomized Clinical Trial. JAMA 2017, 318, 1346–1357. [Google Scholar] [PubMed]
  345. Rhodes, A.; Evans, L.E.; Alhazzani, W.; Levy, M.M.; Antonelli, M.; Ferrer, R.; Kumar, A.; Sevransky, J.E.; Sprung, C.L.; Nunnally, M.E.; et al. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Intensive Care Med. 2017, 43, 304–377. [Google Scholar]
  346. De Backer, D.; Biston, P.; Devriendt, J.; Madl, C.; Chochrad, D.; Aldecoa, C.; Brasseur, A.; Defrance, P.; Gottignies, P.; Vincent, J.L.; et al. Comparison of dopamine and norepinephrine in the treatment of shock. N. Engl. J. Med. 2010, 362, 779–789. [Google Scholar]
  347. Hernandez, G.; Teboul, J.L.; Bakker, J. Norepinephrine in septic shock. Intensive Care Med. 2019, 45, 687–689. [Google Scholar]
  348. Leone, M.; Albanese, J.; Delmas, A.; Chaabane, W.; Garnier, F.; Martin, C. Terlipressin in catecholamine-resistant septic shock patients. Shock. 2004, 22, 314–319. [Google Scholar]
  349. Albanese, J.; Leone, M.; Delmas, A.; Martin, C. Terlipressin or norepinephrine in hyperdynamic septic shock: A prospective, randomized study. Crit. Care Med. 2005, 33, 1897–1902. [Google Scholar]
  350. Gordon, A.C.; Mason, A.J.; Thirunavukkarasu, N.; Perkins, G.D.; Cecconi, M.; Cepkova, M.; Pogson, D.G.; Aya, H.D.; Anjum, A.; Frazier, G.J.; et al. Effect of Early Vasopressin vs Norepinephrine on Kidney Failure in Patients with Septic Shock: The VANISH Randomized Clinical Trial. JAMA 2016, 316, 509–518. [Google Scholar]
  351. Russell, J.A.; Walley, K.R.; Singer, J.; Gordon, A.C.; Hebert, P.C.; Cooper, D.J.; Holmes, C.L.; Mehta, S.; Granton, J.T.; Storms, M.M.; et al. Vasopressin versus norepinephrine infusion in patients with septic shock. N. Engl. J. Med. 2008, 358, 877–887. [Google Scholar]
  352. Tumlin, J.A.; Murugan, R.; Deane, A.M.; Ostermann, M.; Busse, L.W.; Ham, K.R.; Kashani, K.; Szerlip, H.M.; Prowle, J.R.; Bihorac, A.; et al. Outcomes in Patients with Vasodilatory Shock and Renal Replacement Therapy Treated with Intravenous Angiotensin II. Crit. Care Med. 2018, 46, 949–957. [Google Scholar] [PubMed]
  353. Nigwekar, S.U.; Waikar, S.S. Diuretics in acute kidney injury. Semin. Nephrol. 2011, 31, 523–534. [Google Scholar] [PubMed]
  354. Ho, K.M.; Sheridan, D.J. Meta-analysis of frusemide to prevent or treat acute renal failure. BMJ 2006, 333, 420. [Google Scholar]
  355. Ho, K.M.; Power, B.M. Benefits and risks of furosemide in acute kidney injury. Anaesthesia 2010, 65, 283–293. [Google Scholar] [PubMed]
  356. Uchino, S.; Doig, G.S.; Bellomo, R.; Morimatsu, H.; Morgera, S.; Schetz, M.; Tan, I.; Bouman, C.; Nacedo, E.; Gibney, N.; et al. Diuretics and mortality in acute renal failure. Crit. Care Med. 2004, 32, 1669–1677. [Google Scholar]
  357. Mehta, R.L.; Pascual, M.T.; Soroko, S.; Chertow, G.M.; Group, P.S. Diuretics, mortality, and nonrecovery of renal function in acute renal failure. JAMA 2002, 288, 2547–2553. [Google Scholar]
  358. Perazella, M.A. Drug use and nephrotoxicity in the intensive care unit. Kidney Int. 2012, 81, 1172–1178. [Google Scholar]
  359. Morales-Alvarez, M.C. Nephrotoxicity of Antimicrobials and Antibiotics. Adv. Chronic Kidney Dis. 2020, 27, 31–37. [Google Scholar]
  360. Pannu, N.; Nadim, M.K. An overview of drug-induced acute kidney injury. Crit. Care Med. 2008, 36 (Suppl. S4), S216–S223. [Google Scholar]
  361. Perazella, M.A. Renal vulnerability to drug toxicity. Clin. J. Am. Soc. Nephrol. 2009, 4, 1275–1283. [Google Scholar]
  362. Nolin, T.D.; Himmelfarb, J. Mechanisms of drug-induced nephrotoxicity. Handb. Exp. Pharmacol. 2010, 111–130. [Google Scholar]
  363. Bagshaw, S.M.; Wald, R. Strategies for the optimal timing to start renal replacement therapy in critically ill patients with acute kidney injury. Kidney Int. 2017, 91, 1022–1032. [Google Scholar] [PubMed]
  364. Ostermann, M.; Joannidis, M.; Pani, A.; Floris, M.; De Rosa, S.; Kellum, J.A.; Ronco, C.; 17th Acute Disease Quality Initiative (ADQI) Consensus Group. Patient Selection and Timing of Continuous Renal Replacement Therapy. Blood Purif. 2016, 42, 224–237. [Google Scholar] [PubMed]
  365. Bagshaw, S.M.; Wald, R. Indications and Timing of Continuous Renal Replacement Therapy Application. Contrib. Nephrol. 2018, 194, 25–37. [Google Scholar] [PubMed]
  366. Bedford, M.; Stevens, P.; Coulton, S.; Billings, J.; Farr, M.; Wheeler, T.; Kalli, M.; Mottishaw, T.; Farmer, C. Development of risk models for the prediction of new or worsening acute kidney injury on or during hospital admission: A cohort and nested study. Health Serv. Deliv. Res. 2016, 4, 1–8. [Google Scholar]
  367. Park, S.; Cho, H.; Park, S.; Lee, S.; Kim, K.; Yoon, H.J.; Park, J.; Choi, Y.; Lee, S.; Kim, J.H.; et al. Simple Postoperative AKI Risk (SPARK) Classification before Noncardiac Surgery: A Prediction Index Development Study with External Validation. J. Am. Soc. Nephrol. 2019, 30, 170–181. [Google Scholar]
  368. Srisawat, N.; Sileanu, F.E.; Murugan, R.; Bellomod, R.; Calzavacca, P.; Cartin-Ceba, R.; Cruz, D.; Finn, J.; Hoste, E.E.; Kashani, K.; et al. Variation in risk and mortality of acute kidney injury in critically ill patients: A multicenter study. Am. J. Nephrol. 2015, 41, 81–88. [Google Scholar]
  369. Perna, A.F.; Luciano, M.G.; Ingrosso, D.; Pulzella, P.; Sepe, I.; Lanza, D.; Violetti, E.; Capasso, R.; Lombardi, C.; De Santo, N.G. Hydrogen sulphide-generating pathways in haemodialysis patients: A study on relevant metabolites and transcriptional regulation of genes encoding for key enzymes. Nephrol. Dial. Transpl. 2009, 24, 3756–3763. [Google Scholar]
  370. Perna, A.F.; Sepe, I.; Lanza, D.; Ingrosso, D. Hydrogen sulfide increases after a single hemodialysis session. Kidney Int. 2011, 80, 1108–1109. [Google Scholar]
  371. Ostermann, M.; Zarbock, A.; Goldstein, S.; Kashani, K.; Macedo, E.; Murugan, R.; Bell, M.; Forni, L.; Guzzi, L.; Joannidis, M.; et al. Recommendations on Acute Kidney Injury Biomarkers From the Acute Disease Quality Initiative Consensus Conference: A Consensus Statement. JAMA Netw. Open 2020, 3, e2019209. [Google Scholar]
  372. Pickkers, P.; Darmon, M.; Hoste, E.; Joannidis, M.; Legrand, M.; Ostermann, M.; Prowle, J.R.; Schneider, A.; Schetz, M. Acute kidney injury in the critically ill: An updated review on pathophysiology and management. Intensive Care Med. 2021, 47, 835–850. [Google Scholar] [PubMed]
Figure 1. Effect of H2S in pathophysiological AKI: In the absence or at the low physiological level of H2S blood pressure [22,23], oxidative damage [24,25,26], inducible NO synthase [27,28], NF-kB activation [28,42], and apoptosis increase [29,30] and Heme oxygenase 1 decreases [28,31,32] in aging AKI. When H2S is supplemented, these changes are reversed (shown on the right side of the schematic images).
Figure 1. Effect of H2S in pathophysiological AKI: In the absence or at the low physiological level of H2S blood pressure [22,23], oxidative damage [24,25,26], inducible NO synthase [27,28], NF-kB activation [28,42], and apoptosis increase [29,30] and Heme oxygenase 1 decreases [28,31,32] in aging AKI. When H2S is supplemented, these changes are reversed (shown on the right side of the schematic images).
Biomolecules 14 01165 g001
Figure 2. H2S production pathway in a cell. In cytosol, L-Methionine is converted into L-homocysteine by S-Adenosylmethionine synthase (SAM) and S-Adenosyl homocysteine hydrolase (SAH). L-homocysteine is condensed with serine by cystathionine β-synthase (CBS) to generate L-cystathionine, which is converted to L-cysteine by cystathionine γ-lyase (CSE). L-cysteine can be used as a substrate by both CBS and CSE to produce hydrogen sulfide (H2S). CSE catalyzes H2S production by two pathways; one is the conversion of L-homocysteine to L-homolanthionine. Additionally, CSE also catalyzes H2S production by the conversion of L-homocysteine to α-ketoglutarate. L-cysteine is transported into mitochondria, and it is converted by aspartate aminotransferase (AAT) in the presence of α-ketoglutarate to form 3-mercaptopyruvate (3-MP) and L-glutamate. Another enzyme, 3-mercaptopyruvate sulfurtransferase (3-MST), contributes to the endogenous H2S production in the mitochondria by using 3-mercaptopyruvate (3-MP) as a substrate. Peroxisome resident D-amino acid oxidase (DAO) catalyzes D-cysteine into 3-mercaptopyruvate (3-MP), which is then transported into mitochondria and utilized by 3-MST for the production of H2S.
Figure 2. H2S production pathway in a cell. In cytosol, L-Methionine is converted into L-homocysteine by S-Adenosylmethionine synthase (SAM) and S-Adenosyl homocysteine hydrolase (SAH). L-homocysteine is condensed with serine by cystathionine β-synthase (CBS) to generate L-cystathionine, which is converted to L-cysteine by cystathionine γ-lyase (CSE). L-cysteine can be used as a substrate by both CBS and CSE to produce hydrogen sulfide (H2S). CSE catalyzes H2S production by two pathways; one is the conversion of L-homocysteine to L-homolanthionine. Additionally, CSE also catalyzes H2S production by the conversion of L-homocysteine to α-ketoglutarate. L-cysteine is transported into mitochondria, and it is converted by aspartate aminotransferase (AAT) in the presence of α-ketoglutarate to form 3-mercaptopyruvate (3-MP) and L-glutamate. Another enzyme, 3-mercaptopyruvate sulfurtransferase (3-MST), contributes to the endogenous H2S production in the mitochondria by using 3-mercaptopyruvate (3-MP) as a substrate. Peroxisome resident D-amino acid oxidase (DAO) catalyzes D-cysteine into 3-mercaptopyruvate (3-MP), which is then transported into mitochondria and utilized by 3-MST for the production of H2S.
Biomolecules 14 01165 g002
Figure 3. Effects of H2S on renal function. (A) H2S inhibits the activity of tubular Na+-K+-2C channels and Na+/K+-ATPase channels, thereby enhancing renal excretory function, such as RBF and GFR, and the restoration of the oxygen level. (B) H2S as an O2 sensor in the kidney. In normoxic conditions, H2S is metabolized into sulfates in the presence of O2; in hypoxic conditions, the shortage of O2 leads to the accumulation of H2S, which helps to restore the O2 level by enhancing blood flow and suppressing tubular transport activity in the kidney. H2S, hydrogen sulfide; RBF, renal blood flow; GFR, glomerular filtration rate; Una.V, urinary sodium; Uk.V, urinary potassium.
Figure 3. Effects of H2S on renal function. (A) H2S inhibits the activity of tubular Na+-K+-2C channels and Na+/K+-ATPase channels, thereby enhancing renal excretory function, such as RBF and GFR, and the restoration of the oxygen level. (B) H2S as an O2 sensor in the kidney. In normoxic conditions, H2S is metabolized into sulfates in the presence of O2; in hypoxic conditions, the shortage of O2 leads to the accumulation of H2S, which helps to restore the O2 level by enhancing blood flow and suppressing tubular transport activity in the kidney. H2S, hydrogen sulfide; RBF, renal blood flow; GFR, glomerular filtration rate; Una.V, urinary sodium; Uk.V, urinary potassium.
Biomolecules 14 01165 g003
Figure 4. Schematic representation of non-coding RNAs (ncRNAs) that play a role in acute kidney injury (AKI) and subsequent kidney repair. Non-coding RNAs (ncRNAs) are the genome transcripts that function at the RNA level instead of the protein level. AKI, mainly induced by sepsis, renal ischemia-reperfusion, and nephrotoxicity, is pathologically characterized by the injury and death of renal tubular epithelial cells. Complete repair restores the structural integrity and full normal function of the kidney, while maladaptive or incomplete repair leads to chronic pathologies and likely progression to chronic kidney disease (CKD). ncRNAs play regulatory roles in both kidney injury and repair processes, with some being protective and others being pathogenic.
Figure 4. Schematic representation of non-coding RNAs (ncRNAs) that play a role in acute kidney injury (AKI) and subsequent kidney repair. Non-coding RNAs (ncRNAs) are the genome transcripts that function at the RNA level instead of the protein level. AKI, mainly induced by sepsis, renal ischemia-reperfusion, and nephrotoxicity, is pathologically characterized by the injury and death of renal tubular epithelial cells. Complete repair restores the structural integrity and full normal function of the kidney, while maladaptive or incomplete repair leads to chronic pathologies and likely progression to chronic kidney disease (CKD). ncRNAs play regulatory roles in both kidney injury and repair processes, with some being protective and others being pathogenic.
Biomolecules 14 01165 g004
Figure 5. Schematic overview of canonical and non-canonical biogenesis pathways of miRNA: The canonical miRNA pathway produces Pri-miRNA transcripts from miRNA genes by RNA polymerase II, which is processed into pre-miRNAs by the DROSHA/DGCR8 complex. In the non-canonical pathway, pre-miRNAs are directly produced by the splicing of short introns without DROSHA/DGCR8 processing. After transportation to the cytoplasm via Exportin 5, these pre-miRNAs are converted into a double-stranded miRNA/miRNA duplex by DICER. After that, the mature miRNA integrates into RISC. One strand of this duplex is loaded onto AGO and incorporated into the RNA-induced silencing complex (RISC), leading to gene silencing by mRNA degradation and/or translational repression.
Figure 5. Schematic overview of canonical and non-canonical biogenesis pathways of miRNA: The canonical miRNA pathway produces Pri-miRNA transcripts from miRNA genes by RNA polymerase II, which is processed into pre-miRNAs by the DROSHA/DGCR8 complex. In the non-canonical pathway, pre-miRNAs are directly produced by the splicing of short introns without DROSHA/DGCR8 processing. After transportation to the cytoplasm via Exportin 5, these pre-miRNAs are converted into a double-stranded miRNA/miRNA duplex by DICER. After that, the mature miRNA integrates into RISC. One strand of this duplex is loaded onto AGO and incorporated into the RNA-induced silencing complex (RISC), leading to gene silencing by mRNA degradation and/or translational repression.
Biomolecules 14 01165 g005
Figure 6. Two subtypes of macrophages (mφ)-M1 and M2 and their role in AKI. Distinct macrophage phenotypes are involved in renal injury and repair, especially in ischemia-reperfusion injury. Mφ are broadly classified into two subpopulations according to their phenotype and function. So-called “classically activated” pro-inflammatory macrophages (M1) infiltrate the kidney 24 h after reperfusion and contribute to kidney injury. M1 contributes to inflammation by the secretion of cytokines and reactive oxygen species (ROS). M1 also promotes kidney fibrosis through the release of MMP-9. In contrast, “Alternatively” activated anti-inflammatory macrophages (M2) are detected in the kidney 3 days after reperfusion, dampening renal inflammation and promoting tissue repair. M2 macrophages mediate kidney repair by the secretion of Wnt7b, BRP-39, and heme oxygenase-1 (HO-1). Additionally, galectin-3 and TGF-β, released by M2 macrophages, may induce renal fibrosis.
Figure 6. Two subtypes of macrophages (mφ)-M1 and M2 and their role in AKI. Distinct macrophage phenotypes are involved in renal injury and repair, especially in ischemia-reperfusion injury. Mφ are broadly classified into two subpopulations according to their phenotype and function. So-called “classically activated” pro-inflammatory macrophages (M1) infiltrate the kidney 24 h after reperfusion and contribute to kidney injury. M1 contributes to inflammation by the secretion of cytokines and reactive oxygen species (ROS). M1 also promotes kidney fibrosis through the release of MMP-9. In contrast, “Alternatively” activated anti-inflammatory macrophages (M2) are detected in the kidney 3 days after reperfusion, dampening renal inflammation and promoting tissue repair. M2 macrophages mediate kidney repair by the secretion of Wnt7b, BRP-39, and heme oxygenase-1 (HO-1). Additionally, galectin-3 and TGF-β, released by M2 macrophages, may induce renal fibrosis.
Biomolecules 14 01165 g006
Figure 7. Schematic diagram showing some of the current therapeutic options of AKI.
Figure 7. Schematic diagram showing some of the current therapeutic options of AKI.
Biomolecules 14 01165 g007
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

Gupta, S.; Mandal, S.; Banerjee, K.; Almarshood, H.; Pushpakumar, S.B.; Sen, U. Complex Pathophysiology of Acute Kidney Injury (AKI) in Aging: Epigenetic Regulation, Matrix Remodeling, and the Healing Effects of H2S. Biomolecules 2024, 14, 1165. https://doi.org/10.3390/biom14091165

AMA Style

Gupta S, Mandal S, Banerjee K, Almarshood H, Pushpakumar SB, Sen U. Complex Pathophysiology of Acute Kidney Injury (AKI) in Aging: Epigenetic Regulation, Matrix Remodeling, and the Healing Effects of H2S. Biomolecules. 2024; 14(9):1165. https://doi.org/10.3390/biom14091165

Chicago/Turabian Style

Gupta, Shreyasi, Subhadeep Mandal, Kalyan Banerjee, Hebah Almarshood, Sathnur B. Pushpakumar, and Utpal Sen. 2024. "Complex Pathophysiology of Acute Kidney Injury (AKI) in Aging: Epigenetic Regulation, Matrix Remodeling, and the Healing Effects of H2S" Biomolecules 14, no. 9: 1165. https://doi.org/10.3390/biom14091165

APA Style

Gupta, S., Mandal, S., Banerjee, K., Almarshood, H., Pushpakumar, S. B., & Sen, U. (2024). Complex Pathophysiology of Acute Kidney Injury (AKI) in Aging: Epigenetic Regulation, Matrix Remodeling, and the Healing Effects of H2S. Biomolecules, 14(9), 1165. https://doi.org/10.3390/biom14091165

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