Abstract
Acute kidney injury (AKI) is a major public health concern associated with high morbidity and mortality. Despite decades of research, the pathogenesis of AKI remains incompletely understood and effective therapies are lacking. An increasing body of evidence suggests a role for epigenetic regulation in the process of AKI and kidney repair, involving remarkable changes in histone modifications, DNA methylation and the expression of various non-coding RNAs. For instance, increases in levels of histone acetylation seem to protect kidneys from AKI and promote kidney repair. AKI is also associated with changes in genome-wide and gene-specific DNA methylation; however, the role and regulation of DNA methylation in kidney injury and repair remains largely elusive. MicroRNAs have been studied quite extensively in AKI, and a plethora of specific microRNAs have been implicated in the pathogenesis of AKI. Emerging research suggests potential for microRNAs as novel diagnostic biomarkers of AKI. Further investigation into these epigenetic mechanisms will not only generate novel insights into the mechanisms of AKI and kidney repair but also might lead to new strategies for the diagnosis and therapy of this disease.
Acute kidney injury (AKI), characterized by a rapid decline in kidney function, is a major public health problem. It is responsible for approximately 1.7 million deaths per year worldwide and is associated with increased length of hospital stay among hospitalized patients as well as high costs1–3. The pathophysiology of AKI is incompletely understood but involves the injury and death of renal tubular cells, particularly of cells in the proximal tubule4,5. Following injury, a repair response is activated, which involves epithelial cell de-differentiation, proliferation and re-differentiation. However, severe or sustained injury often results in maladaptive and incomplete repair, leading to tubular degeneration, inflammation, renal fibrosis and ultimately progression to chronic kidney disease (CKD) or end-stage renal disease 6–10 (FIG. 1). Currently, other than supportive care in the form of dialysis, no effective treatments for AKI are available.
Fig. 1 |. Pathophysiology of AKI and repair.
The pathophysiology of acute kidney injury (AKI) is very complex, involving interplay between tubular, microvascular and inflammatory factors. Acute injury insults typically induce the injury and death of tubular epithelial cells, injury and activation of endothelial cells and leukocyte infiltration, culminating in renal dysfunction. In the presence of mild injury, adaptive repair mechanisms can restore epithelial integrity, suppress the immune response and re-establish healthy vasculature. By contrast, severe or persistent injury induces maladaptive repair. Tubular cells may undergo G2/M cell cycle arrest, senescence and apoptosis or necrosis, leading to the release of profibrotic and pro-inflammatory factors. Tubular atrophy and degeneration, together with sustained inflammation and microvascular loss, result in renal interstitial fibrosis, characterized by the proliferation and activation of fibroblasts and deposition of extracellular matrix (ECM), ultimately leading to chronic kidney disease (CKD).
Epigenetics is the study of the heritable mechanisms that control gene expression without changing the primary nucleotide sequence. Epigenetic mechanisms, including histone modifications, DNA methylation and non-coding RNAs, can induce changes to a phenotype but cannot change a genotype. In general, epigenetic modifications are considered to be stable and heritable during cell divisions; however, they are potentially reversible and can be affected by environmental factors, age and disease state. Emerging evidence suggests that epigenetic regulation contributes to various kidney diseases, including diabetic kidney disease, CKD and renal cell carcinoma. For example, epigenetic alterations commonly occur in CKD, associated with genes involved in fibrosis, inflammation and epithelial-to-mesenchymal transition11,12. In particular, global targeting or gene-specific targeting of DNA methylation has been reported to effectively inhibit CKD progression in animal models of disease, suggesting that targeting DNA methylation could be a new therapeutic approach for the treatment of CKD13–17. An accumulating body of evidence also suggests a crucial role for epigenetic mechanisms in AKI and repair. In this Review, we provide a general overview of the main epigenetic mechanisms that have been linked to AKI, describe available evidence linking epigenetic changes to kidney injury and repair in AKI and discuss the challenges and therapeutic implications of these findings.
Epigenetic mechanisms
Epigenetic regulation involves the covalent modification of DNA or histone proteins, or RNA interference by non-coding RNAs, to modulate gene expression. Modification of DNA or histone proteins is achieved through DNA methylation or a number of histone modifications in processes that are catalysed by specific enzymes called epigenetic writers. These modifications are then recognized by epigenetic readers and can be removed by epigenetic erasers.
Histone modifications
Histones, including core histones (H2A, H2B, H3 and H4) and linker histones (H1 and H5), are highly conserved, basic or positively charged proteins that are involved in DNA packaging. They can associate with negatively charged DNA through electrostatic interactions and package it into highly condensed and ordered chromatin structure units called nucleosomes18. Each nucleosome consists of a segment of DNA wrapped around a core histone octamer, which contains two copies of each core histone protein (that is, two H2A–H2B dimers and one H3–H4 tetramer)19,20. Histone modifications involve the covalent, post-translational modifications of core histone proteins and include, but are not limited to, acetylation, methylation, phosphorylation, ubiquitylation, sumoylation, citrullination, biotinylation, crotonylation and ADP ribosylation. These modifications occur predominantly, but not exclusively, on the amino-terminal tails of the histones and are thought to change the structure of chromatin or provide docking sites for transcriptional regulators to either positively or negatively regulate gene expression19,20. Acetylation, methylation and phosphorylation are the major forms of histone modification (FIG. 2).
Fig. 2 |. Mechanisms and consequences of histone modifications.
a | Histone modifications (acetylation, methylation and phosphorylation) are catalysed by specific enzymes known as epigenetic writers, recognized by epigenetic readers and removed by epigenetic erasers. b | The major sites of histone acetylation, methylation and phosphorylation. Generally, histone acetylation is associated with permissive transcription, and histone methylation is associated with either active (H3 lysine 4 (H3K4), H3K36 and H3K79) or repressive (H3K9, H3K27 and H4K20) transcription. Phosphorylation of histones (for example, phosphorylation of H3 at serine 10 (H3S10ph), threonine 11 (H3T11ph), serine 28 (H3S28ph) and tyrosine 41 (H3Y41ph) and phosphorylation of H2B at serine 32 (H2BS32ph)) is often associated with transcriptional activation; however, histone phosphorylation (for example, of H2A at serine 1 (H2AS1ph), phosphorylation of H2B at tyrosine 37 (H2BY37ph) and phosphorylation of H4 at serine 1 (H4S1ph)) can be associated with transcriptional repression. HATs, histone acetyltransferases; HDACs, histone deacetylases; HDMs, histone demethylases; HMTs, histone methyltransferases.
Histone acetylation
Histone acetylation involves the addition of a negatively charged acetyl group to the lysine of core histones by histone acetyltransferases (HATs) and is one of the most extensively studied forms of histone modification. Histone acetylation neutralizes the positive charge of lysines, promoting an open chromatin configuration and facilitating access of transcription factors, leading to permissive transcription. Moreover, acetylated histone residues serve as docking sites for many transcriptional co-activators that promote transcription21. Histone acetylation is reversible and deacetylation is catalysed by histone deacetylases (HDACs). To date, 18 mammalian HDAC proteins have been identified, which, on the basis of their domain organization and sequence homology to yeast orthologues, are divided into four classes: class I (HDACs 1, 2, 3 and 8), class II (HDACs 4, 5, 6, 7, 9 and 10), class III (sirtuin (SIRT) 1–7) and class IV (HDAC11). Classes I, II and IV require zinc for their catalytic activity and are considered to be the ‘classical’ HDACs, whereas class III HDACs depend on NAD+ for their catalytic activity22.
Histone methylation
Histone methylation involves the addition of a methyl group to a basic amino acid on core histones in a process mediated by histone methyltransferases (HMTs). Any of the three basic amino acids can be methylated, but it most commonly occurs on lysine and arginine residues and rarely at histidine residues. Lysine and arginine can be subjected to multiple methylations on their side chains. Specifically, lysine can be monomethylated, dimethylated or trimethylated on its ε-amine group and arginine can be monomethylated, or symmetrically or asymmetrically dimethylated, on its guanidyl group23. Unlike histone acetylation, histone methylation does not change the charge of the histone protein but rather provides sites for the docking of transcription regulators. Histone methylation is associated with either active or repressive transcription, depending on the amino acids methylated and the number of methyl groups attached. Generally, histone methylation at H3 lysine 4 (H3K4), H3K36 and H3K79 activates gene transcription, whereas methylation at H3K9, H3K27 and H4K20 is associated with repression of transcription24. Histone methylation was once thought to be permanent; however, the identification of novel histone demethylases, such as lysine-specific demethylases (LSD1 and LSD2) and Jumonji (JmjC)-domain containing histone demethylases, has challenged this view25.
Histone phosphorylation
Histone phosphorylation is another major form of histone modification that changes the chromatin structure. It can take place on all four core histones, mainly on serine, threonine and tyrosine residues. Phosphorylation of different core histones requires distinct protein kinases, such as MEC1 for the phosphorylation of H2A in yeast, MST1 (mammalian sterile-20-like kinase) for the phosphorylation of H2B in mammals, the Aurora kinases for the phosphorylation of H3 and SPS1 (a sporulation-specific kinase) for the phosphorylation of H4 in yeast20. Phosphorylation of histones is often associated with transcriptional activation (for example, phosphorylation of H3 at serine 10 (H3S10ph), threonine 11 (H3T11ph), serine 28 (H3S28ph) and tyrosine 41 (H3Y41ph) and phosphorylation of H2B at serine 32 (H2BS32ph)); however, it can sometimes be associated with transcriptional repression (for example, phosphorylation of H2A at serine 1 (H2AS1ph), phosphorylation of H2B at tyrosine 37 (H2BY37ph) and phosphorylation of H4 at serine 1 (H4S1ph))26–28. Increasing evidence indicates that histone phosphorylation not only is involved in the regulation of gene transcription but also is involved in the DNA damage repair response and chromatin condensation during mitosis and meiosis26.
Histone modifications in AKI and repair
Histone modifications, particularly histone acetylation, are among the most studied epigenetic mechanisms in AKI and repair. Improved understanding of the contribution of these modifications to the process of injury and repair might open new therapeutic avenues to prevent injury or promote repair.
Histone acetylation in AKI and repair
An accumulating body of evidence suggests that AKI is associated with changes in histone acetylation, especially in the context of ischaemic AKI. A 2008 study showed dynamic changes in histone acetylation in AKI induced by renal ischaemia–reperfusion injury (IRI)29. In this model, severe unilateral ischaemia induced a transient reduction in H3 acetylation in proximal tubular cells, probably resulting from a decrease in HAT activity. During reperfusion, however, H3 acetylation was restored and expression of bone morphogenetic protein 7 (BMP7) — a key regulator of renal repair — was induced, at least in part owing to selective downregulation of HDAC5. In contrast to the findings from this study, another study using a model of moderate unilateral IRI reported an increase in H3 acetylation after 1 day of reperfusion that persisted for 3 weeks30. Moreover, another study in a mouse model of bilateral IRI reported an initial increase in acetylation of H3K14 (H3K14Ac) followed by a decrease to basal levels31. By contrast, H4K5Ac and H4K12Ac were reduced and then restored. Dynamic regulation of the HBO1 (also known as KAT7, an H4-specific HAT)–JADE1 complex, was found to contribute to changes in histone H4 acetylation31. Although these studies demonstrate that dynamic alterations in histone acetylation occur in the context of AKI, inconsistencies in the direction of change require clarification. Several possible reasons exist for the differences in histone acetylation observed between the different studies, including differences in the severity of injury and mouse strain and the use of different time points and methods for the analysis of histone acetylation.
Several other studies have examined the role of histone acetylation on the expression of genes involved in ischaemic AKI. One study demonstrated that upregulation of the rate-limiting enzyme in cholesterol synthesis, high mobility group CoA reductase (HMG-CoA reductase; encoded by HMGCR), contributed to cholesterol loading and cytoprotective effects in ischaemic AKI32. Interestingly, the histone variant H2A.Z and histone modifications (trimethylation at H3K4 (H3K4m3) and H3K19Ac) in the Hmgcr promoter and along the gene were increased and likely contributed to the upregulation of HMG-CoA reductase expression32. This hypothesis is supported by the finding that compared with patients without AKI, patients with AKI had increased HMG-CoA reductase activity associated with increased H3K4m3 at HMGCR33. The transcriptional repressor ATF3 (activating transcription factor 3) has renoprotective effects in ischaemic AKI by down-regulating the expression of inflammatory cytokines, such as IL-6 and IL-12B34. Induction of ATF3 in a mouse model of ischaemic AKI led to the recruitment of HDAC1 to the promoter regions of Il6 and Il12b, resulting in the suppression of Il6 and Il12b expression and inhibiting the inflammatory response. Silencing of HDAC1 exacerbated AKI, associated with higher expression of IL-6 and IL-12B. In another study, upregulation of plasminogen activator inhibitor type 1 (PAI1; in mice encoded by the Serpine1 gene (serine (cysteine) pepti-dase inhibitor, clade E, member 1)), which contributes to the susceptibility of male mice to ischaemic AKI, was associated with a decrease in HDAC11 expression and increased acetylation of H3 in the Serpine1 promoter35.
In addition to renal IRI, histone acetylation is involved in other forms of AKI. For example, decreased H3 acetylation has been reported in both in vivo and in vitro experimental models of septic AKI, and administration of the α2-adrenoceptor agonist dexmedetomidine protected mice against septic AKI by inhibiting HDAC2 and HDAC5 to induce H3 acetylation and the expression of BMP7 (REF.36). In a mouse model of lipopolysaccharide (LPS)-induced septic AKI, upregulation of p300/CBP-associated factor (PCAF; a HAT) was associated with increased acetylation of histones, such as H3K18Ac, and increased expression of inflammatory genes37. Silencing of PCAF in cultured renal tubular cells significantly reduced H3K18Ac, resulting in decreased expression of inflammatory factors, such as vascular cell adhesion molecule 1 (VCAM1), intercellular adhesion molecule 1 (ICAM1) and monocyte chemoattractant 1 (MCP1; also known as CCL2), suggesting that induction of H3K18Ac contributes to the upregulation of inflammatory genes in septic AKI. Peroxisome proliferator-activated receptor-γ co-activator 1α (PGC1α; in mice encoded by the Ppargc1a gene) is a regulator of mitochondrial biogenesis that is protective in AKI and is downregulated in several experimental models of AKI38–40. A 2016 study showed that suppression of PGC1α in mice with folic-acid-induced AKI was mediated through the actions of a cytokine called TWEAK (TNF-related weak inducer of apoptosis), which promoted H3 deacetylation at the Ppargc1a promoter, leading to chromatin condensation and repression of PGC1α expression41. Moreover, downregulation of another renoprotective factor, Klotho (in mice encoded by Kl), in folic acid-induced AKI was also shown to occur via TWEAK-mediated deacetylation of the Kl gene promoter42. In rats with cisplatin-induced AKI, induction of the class III HDAC protein SIRT1 occurred as early as 6 h after cisplatin administration and was associated with deacetylation of histone H3, although expression of HDAC5 and HDAC3 were not induced until days 2 and 3, respectively43. Overexpression of SIRT1 protects against cisplatin-induced AKI by retaining peroxisome function44. The protective effects of SIRT1 are also supported by the finding that suppression of SIRT1 reduced resistance of cultured renal medullary interstitial cells to oxidative stress and that SIRT1 deficiency aggravated renal apoptosis and fibrosis in mice with unilateral ureteral obstruction (UUO)45. Taken together, these studies support an important role of histone acetylation in various forms of AKI.
Histone acetylation-based drugs in AKI and repair.
As described above, histone acetylation, which is regulated by the balance between HATs and HDACs, has a key role in chromatin remodelling and therefore in the regulation of gene expression. The involvement of histone acetylation in AKI and repair suggests that HDAC inhibitors (HDACis), HDAC activators or HAT inhibitors might have therapeutic value (TABLE 1).
Table 1|.
Histone-acetylation-based epigenetic drugs in AKI and repair
| Drug | Model | Function | Mechanisms | Refs |
|---|---|---|---|---|
| HDACis | ||||
| TSA | Cisplatin nephrotoxicity in immortalized mouse proximal tubule cells | Protective | Restores CREB-mediated transcription as well as survival | 47 |
| Cisplatin nephrotoxicity in RPTCs | Protective | Delays p53 phosphorylation, acetylation and activation | 48 | |
| Mouse cisplatin nephrotoxicity | Protective | Upregulates AMWAP | 49 | |
| Mouse cisplatin nephrotoxicity | Protective | Activates autophagy | 50 | |
| Mouse UUO | Protective | Enhances H3 and H4 acetylation and inhibits STAT3 activation | 53 | |
| Mouse unilateral IRI | Protective | Enhances H3 acetylation and miR-21 | 54 | |
| SAHA | Cisplatin nephrotoxicity in RPTCs | Protective | Inhibits p53 phosphorylation, acetylation and activation and blocks CHK2 phosphorylation | 48 |
| Rat haemorrhagic shock | Protective | Increases H3K9Ac and decreases BAD | 55 | |
| VPA | Rat haemorrhagic shock | Protective | Increases H3K9Ac and BCL-2 and decreases BAD | 55 |
| Mouse adriamycin-induced nephropathy | Protective | Increases glomerular H3K9Ac | 56 | |
| Rat IRI | Protective | NA | 57 | |
| MS-275 | Mouse cisplatin nephrotoxicity | Protective | Upregulates AMWAP | 49 |
| Mouse unilateral IRI | Protective | Enhances H3 acetylation | 54 | |
| Mouse folic-acid-induced AKI | Pathogenic | Increases H3 acetylation, suppresses the expression and phosphorylation of EGFR and inhibits STAT3 and AKT phosphorylation | 58 | |
| Mouse rhabdomyolysis-induced AKI | Pathogenic | Increases H3 acetylation and suppresses the expression and phosphorylation of EGFR | 58 | |
| m4PTB | Zebrafish gentamicin-induced AKI | Protective | Increases proliferation of tubular epithelial cells | 59 |
| Mouse IRI | Protective | Increases proliferation and reduces G2/M arrest of renal tubular epithelial cells and prevents interstitial fibrosis | 59 | |
| Mouse aristolochic acid nephropathy | Protective | Increases proliferation and reduces G2/M arrest of renal tubular epithelial cells and prevents interstitial fibrosis | 60 | |
| Tubastatin A | Mouse rhabdomyolysis-induced AKI | Protective | Increases H3 acetylation and inhibits phosphorylation of NF-κB, expression of multiple cytokines and chemokines and oxidative stress | 61 |
| Mouse cisplatin nephrotoxicity | Protective | Increases H3 acetylation, phosphorylation of AKT and autophagy and inhibits phosphorylation of NF-κB, expression of multiple cytokines and chemokines and oxidative stress | 62 | |
| SIRT1 (HDAC) activators | ||||
| Resveratrol | Cultured RMICs treated with H2O2 | Protective | NA | 45 |
| Rat cisplatin nephrotoxicity | Protective | Inhibits inflammatory cell filtration and reduces lipid peroxidation and glutathione depletion | 63 | |
| Rat septic AKI | Protective | Deacetylates SOD2 | 64 | |
| SRT-2183 | Cultured RMICs treated with H2O2 | Protective | Induces COX2 and PGE2 | 45 |
| SRT-1720 | Mouse UUO | Protective | Induces COX2 and PGE2 | 45 |
| Mouse IRI | Protective | NA | 65 | |
| HAT inhibitor | ||||
| Curcumin | Rat cisplatin nephrotoxicity | Protective | Inhibits inflammatory response and oxidative stress | 66 |
| Rat IRI | Protective | Reduces oxidation products MDA, NO and PC in serum and kidney tissue | 67 | |
| Rat LPS-induced AKI | Protective | Inhibits interstitial inflammatory infiltration | 68 | |
AKI, acute kidney injury; AKT, protein kinase B; AMWAP, activated microglia/macrophage WAP domain protein; BAD, BCL-2 associated death promoter protein; BCL-2, B cell lymphoma 2; CHK2, checkpoint kinase 2; COX2, cyclooxygenase 2; CREB, cAMP-responsive element binding protein; EGFR, epidermal growth factor receptor; H3K9Ac, histone H3 lysine 9 acetylation; HAT, histone acetyltransferase; HDAC, histone deacetylase; HDACi, HDAC inhibitor; IRI, ischaemia–reperfusion injury; LPS, lipopolysaccharide; m4PTB, 4-methyl-thiobutanate; MDA, malondialdehyde; NA, not available; NO, nitric oxide; PC, protein carbonyl; PGE2, prostaglandin E2; RMICs, mouse renal medullary interstitial cells; RPTCs, rat proximal tubular cells; SAHA, suberoylanilide hydroxamic acid; SIRT1, sirtuin 1; SOD2, superoxide dismutase 2; STAT3, signal transducer and activator of transcription 3; TSA, trichostatin A; UUO, unilateral ureteral obstruction; VPA, valproic acid.
The effects of HDACis on AKI were first investigated in the experimental models of cisplatin-induced nephrotoxicity, with conflicting findings46,47. We showed that the HDACis trichostatin A (TSA) and suberoylanilide hydroxamic acid (SAHA) were toxic to renal tubular cells46, whereas other researchers found TSA to exert cytoprotective effects against cisplatin-induced apoptosis in renal tubular cells47. Our follow-up study showed that the effects of both TSA and SAHA on renal tubular cells are dose-dependent: they are toxic at high-micromolar concentrations, but at concentrations <1 μmol/l, they can protect against cisplatin-induced cell death48. In mice with cisplatin-induced AKI, the renoprotective effects of low-dose TSA involve upregulation of an anti-inflammatory protein called activated microglia/macrophage WAP domain protein (AMWAP)49. Our most recent work showed that the protective effect of TSA in cisplatin-induced AKI is diminished by blocking renal tubular autophagy50, suggesting a critical role for autophagy in mediating the protective effect of this HDACi and supporting previous work showing that tubular cell autophagy is protective in AKI51,52. TSA has also been investigated in other models of AKI. A 2009 study demonstrated that TSA ameliorated renal tubular cell apoptosis, caspase 3 activation and renal fibrosis in a mouse model of UUO53. Similarly, TSA protected against ischaemic AKI and consequent interstitial fibrosis in a mouse model of unilateral renal IRI54. Moreover, the HDACi SAHA protected kidney cells from apoptosis induced by lethal haemorrhagic shock in rats, associated with increased H3K9 acetylation55. Valproic acid (VPA), a compound that has HDACi properties, attenuated proteinuria and kidney injury in models of adriamycin-induced nephropathy and IRI, respectively56,57. Similar to the effects of TSA, MS-275, an inhibitor of HDAC1 and HDAC3, attenuated kidney injury and the development of renal fibrosis in models of cisplatin-induced AKI and ischaemic AKI49,54. In models of folic acid and rhabdomyolysis-induced AKI, however, MS-275 exacerbated kidney injury and impaired renal regeneration58. Studies have also assessed the effects of the HDACi 4-methyl-thiobutanate (m4PTB), an analogue of 4-(phenylthio) butanoic acid. Studies of zebrafish and mouse models of AKI suggest that administration of m4PTB promotes recovery following injury59,60. In mouse models of IRI and aristolochic-acid-induced AKI, treatment with m4PTB enhanced proliferation and reduced G2/M arrest of renal tubular epithelial cells and prevented the development of interstitial fibrosis59,60. In addition, inhibition of HDAC6 by tubastatin A enhanced H3 acetylation, suppressed inflammation and oxidative stress and prevented the development of rhabdomyolysis-induced and cisplatin-induced AKI61,62.
In addition to HDACis, the effects of HDAC activators — particularly the SIRT1 activators, resveratrol, SRT-2183 and SRT-1720 — on AKI and subsequent repair have been investigated. A 2010 study reported that resveratrol and SRT-2183 enhanced resistance of primary cultured mouse renal medullary interstitial cells to oxidative stress-induced apoptosis and that administration of SRT-1720 attenuated apoptosis and fibrosis in mice with UUO45. The renoprotective effects of resveratrol have also been demonstrated in experimental models of cisplatin-induced nephrotoxicity and septic AKI63,64. Administration of SRT-1720 to mice with IRI induced proliferation of renal tubular cells and attenuated renal injury, whereas SIRT1 deficiency had the opposite effects, further supporting a role for SIRT1 in the regulation of kidney injury and repair65.
Several studies have examined the role of curcumin, an inhibitor of the HAT PCAF in AKI. In rats, administration of curcumin showed renoprotective effects in various AKI models, including cisplatin-induced nephrotoxicity, IRI and LPS-induced AKI66–68.
Together, these studies support the functional relevance of histone acetylation in AKI and repair. In general, pharmacological inhibition of HDACs seems to protect the kidney against multiple forms of AKI and promotes kidney repair. However, the role of histone acetylation in AKI is controversial (TABLE 1). As described above, HDAC activators, particularly activators of SIRT1, and the HAT inhibitor curcumin also have renoprotective effects, complicating interpretation of the role of histone acetylation in kidney injury and repair. Specific HATs and HDACs might have distinct roles in the pathogenesis of AKI. To date, pan-HDACis and class-specific HDACis have mainly been used to study the role of histone acetylation in AKI and repair. Future studies will need to elucidate the function of specific HDAC isoforms in AKI and repair and develop isoform-selective HDACis for research and therapeutic use. Of note, in addition to changing the acetylation status of histones, HATs and HDACs can acetylate and deacetylate non-histone proteins. For example, p53, a tumour suppressor protein that regulates the cell cycle and has many functions, including a pathogenic role in AKI, is a target of both the HAT PCAF and the HDACs HDCA1 and SIRT1 (REF69). In a model of ischaemic AKI, SIRT1 deficiency increased acetylation and expression of p53, which contributed to the development of kidney injury65. Inhibition of PCAF or activation of SIRT1 might result in the deacetylation and inactivation of p53, suggesting that the renoprotective effects of HDACis, HDAC activators or HAT inhibitors in AKI might involve alterations in the acetylation status of non-histone proteins. However, this proposal requires verification. Identification of the targets of specific HDACs of interest is important to delineate the underlying mechanisms of their roles in the pathogenesis of AKI and could lead to the discovery of novel strategies for the prevention and treatment of AKI.
Other histone modifications
Compared with histone acetylation, the role of histone methylation in AKI and kidney repair has been much less studied. However, induction of inflammatory factors (TNF and MCP1) and HMG-CoA reductase is associated with increased levels of H3K4m3 in various mouse models of AKI32,70,71. Interestingly, urinary levels of MCP1 and H3K4m3 specifically associated with MCP1 and HMGCR were elevated in patients with AKI, suggesting these methylation changes might have potential as biomarkers of AKI33,71. Upregulation of enhancer of zeste homologue 2 (EZH2) — a histone methyltransferase that specifically mediates trimethylation of lysine 27 on histone H3 (H3K27m3) — has been documented in fibrotic kidneys from mice with UUO and in patients with CKD, suggesting that this methyltransferase has profibrotic functions72,73. Pharmacological inhibition of EZH2 decreased H3K27m3 and attenuated renal fibrosis in mice with IRI by suppressing the expression of genes encoding profibrotic proteins, such as collagen type 3a1 (COL3A1) and tissue inhibitor of metalloproteinase 2 (TIMP2)74.
Knowledge of the role of histone phosphorylation in AKI is also very limited. A 2001 study demonstrated that histone H3 phosphorylation induced chromatin condensation and promoted the damage and death of renal proximal tubular cells in response to oxidative stress75. Prevention of H3 phosphorylation by inhibiting MAPK kinase (with the selective MAPK inhibitor PD98059) or by inhibiting poly(ADP-ribose) polymerase (PARP) (with 3-aminobenzamide) promoted renal tubular cell survival75. Phosphorylation of the histone H2A variant H2AX at serine 139, indicative of double-strand DNA damage, has been linked to AKI in various models, including cisplatin-induced AKI and renal IRI76–79. No information is available to date regarding the potential role of histone phosphorylation in kidney repair.
Histone crotonylation is another histone modification that involves the addition of a crotonyl group to lysine residues on core histones by the actions of histone crotonyltransferases. Similar to histone acetylation, histone crotonylation neutralizes the positive charge of lysine residues and stimulates transcription; however, the mechanisms governing crotonylation versus acetylation remain largely unclear80. A 2016 study demonstrated an increase in histone crotonylation in kidney tissues in models of AKI induced by folic acid and cisplatin81. Further analysis indicated that histone crotonylation increased the expression of PGC1α and SIRT3, promoting protective pathways.
Obviously, multiple forms of histone modifications regulate gene expression under pathophysiological conditions, including AKI. A 2015 study mapped patterns of histone modifications in relation to changes in the expression of several genes induced by three models of AKI: unilateral renal IRI, LPS-induced AKI and combined IRI and LPS-induced AKI82. Using matrix chromatin immunoprecipitation, the researchers determined levels of key permissive histone marks (that is, H3K9Ac, H3K18Ac, H3K27Ac, H4K5/8/12/16Ac, H3K4m3, H3K4m2 and H3S10ph), elongation marks (H3K36m3, H3K36m2 and H3K79m2) and repressive histone marks (H3K27m3, M3K9m2, macroH2A, H2AK119ub1, H2BK120ub1 and H4S1ph) at genes that encode the injury markers NGAL and KIM1 and the inflammatory factors TNF and ICAM1. In general, the permissive histone marks were increased and the repressive marks were decreased in these AKI models, indicating that these epigenetic modifications might be responsible for the increased expression of these genes in the context of kidney injury. Unexpectedly, however, histone modifications exhibited striking heterogeneity at different histone marks, in different genes and in different AKI models, and exhibited time-dependent alterations. These findings highlight the complex regulation of histone modifications in AKI and the need to better understand the mechanisms of epigenetic regulation in this disease.
DNA methylation
DNA methylation is the typical epigenetic modification that affects DNA. 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)83,84. DNA methylation occurs predominantly in CpG dinucleotides; however, DNA methylation can also occur at low frequency in non-CpG sites, particularly in embryonic stem cells85,86, oocytes87 and brain tissues88,89. In the mammalian genome, the majority (70–80%) of CpG sites are methylated. CpG islands within promoter regions of genes are usually unmethylated, enabling transcription. In mammals, DNA methylation is critical for the regulation of many biological processes, including embryonic development, genomic integrity, X chromosome inactivation (in females) and genomic imprinting; DNA methylation has also been linked to complex human diseases, including cancer90–92.
The machinery involved in mammalian DNA methylation consists of three parts: DNA methylation writers, which establish and maintain the methylation marks in the genome; DNA methylation readers, which read or interpret the methylation marks; and DNA methylation erasers, which mediate DNA demethylation.
The most notable function of DNA methylation is transcriptional regulation (FIG. 3a). Generally, low levels of DNA methylation (hypomethylation) at a gene promoter correlate with transactivation of that gene through increased binding of transcription factors. Conversely, elevated DNA methylation (hypermethylation) in a gene promoter region typically leads to loss of gene expression93–95. DNA methylation can suppress gene expression by either directly preventing transcription factors from binding the promoter or indirectly through the actions of DNA methylation readers, which bind to methylated CpGs and stimulate the recruitment of repressor complexes.
Fig. 3 |. Mechanisms of DNA methylation and demethylation.
a | DNA methylation involves the covalent addition of a methyl group (CH3) to cytosine by DNA methyltransferases (DNMTs). Initial DNA methylation patterns are established by DNMT3a and DNMT3b. When DNA replication occurs, DNMT1 faithfully copies the DNA methylation patterns from parental strands to daughter strands. DNA methylation can inhibit gene expression directly by preventing transcription factor binding (1) or indirectly through the actions of DNA methylation readers, such as methyl-CpG-binding domain (MBD) proteins, which can either inhibit the binding of transcription factors (2) or recruit transcription repressors (3), leading to gene suppression. b | The process of DNA demethylation can be passive or active. Passive demethylation occurs in the absence of functional DNMT1 during cell division, as newly synthesized DNA strands are not able to be methylated. Methyl groups can also be actively removed from DNA by an enzymatic replacement mechanism called active demethylation. On one hand, 5-methylcytosine (5mC; a methylated form of cytosine) can be deaminated to thymine by activation-induced cytidine deaminase (AID), which generates a mismatch between thymine and guanine bases. 5mC can also be oxidized by ten-eleven translocation (TET) proteins to 5-hydroxymethylcytosine (5hmC), which can be further oxidized to 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) or deaminated to 5-hydroxymethyl-uracil (5hmU). Finally, these intermediates are replaced by cytosine through thymine DNA glycosylase (TDG) and base excision repair (BER). SAH, S-adenosyl-L-homocysteine; SAM, S‑adenosyl-L-methionine.
DNA methylation writers
The DNMT family includes DNMT1, DNMT2 and DNMT3 (DNMT3a, DNMT3b and DNMT3-like (DNMT3L)). DNMT1, DNMT3a and DNMT3b are the major DNMTs that are required for DNA methylation and are conserved in mammals and plants (FIG. 3a). DNMT1 is the maintenance DNMT and has a preference for hemimethylated DNA. During cell division, the newly synthesized daughter DNA strands are recognized by DNMT1 and are methylated faithfully according to the parental DNA strands. DNMT1 is the most abundant DNMT in most cell types, especially in proliferating cells. By contrast, DNMT3a and DNMT3b are de novo DNMTs, which establish the initial DNA methylation patterns. DNMT3a and DNMT3b are predominantly expressed in embryonic tissues with low expression in somatic tissues96. DNMT3L is not an active enzyme but can facilitate the catalytic activity of DNMT3a and DNMT3b by enhancing the binding of the methyl donor, SAM97. DNMT2, also known as transfer RNA aspartic acid methyltransferase 1 (TRDMT1), has barely detectable methylation activity on DNA but has a robust methylation activity on tRNAs, especially at the 38th cytosine residue in the anticodon loop of aspartic acid, glycine and valine tRNAs98,99. Methylation of tRNAs enhances their stability and protects them from stress-induced cleavage99. Deficiency of DNMT1 or DNMT3b is embryonic lethal in mice, whereas mice that are deficient in DNMT3a die within a few weeks of birth, indicating that these DNMTs are essential for animal development100,101.
DNA methylation readers
Three families of DNA methylation readers exist: the methyl-CpG-binding domain (MBD) protein family, the zinc-finger proteins and the ubiquitin-like with PHD and ring finger domain-containing (UHRF) protein family. The MBD family consists of five members: methyl-CpG-binding protein 2 (MeCP2) and MBD1–MBD4 (Refs102,103). Except for MBD4, the MBDs bind to methylated cytosine and repress transcription by recruiting various repressor complexes through their transcriptional repression domain103. For example, MeCP2 binds a single symmetrically methylated CpG site and recruits the transcriptional co-repressors Sin3a (SIN3 transcription regulator family member A) and HDACs104; MBD2 binds to methylated DNA and interacts with MeCP1, a histone deacetylase complex, to inhibit transcription105. The zinc-finger proteins include ZBTB4, ZBTB38 and transcriptional regulator Kaiso (ZBTB33). ZBTB4 and ZBTB38 act as transcription repressors and repress transcription in a methyl-CpG-dependent manner. Kaiso recruits the repressor complexes and suppresses transcription102,106. The UHRF family consists of UHRF1 and UHRF2. Unlike most of the MBD family and the zinc-finger proteins, the UHRF family does not directly repress transcription but recruits DNMT1 to the hemimethylated DNA to maintain DNA methylation103.
DNA methylation erasers
DNA methylation was traditionally considered to be a very stable modification inherited by daughter cells; however, studies over the past decade have demonstrated that DNA methylation is not permanent and can be erased by demethylation, which can occur via passive or active mechanisms (FIG. 3b). Passive DNA demethylation can occur during cell division if DNMT1 is missing or inhibited. Under these circumstances, the newly synthesized DNA strands remain unmethylated, leading to passive demethylation in the daughter cells. Conversely, active demethylation can be achieved in both dividing and non-dividing cells by DNA methylation erasers such as activation-induced cytidine deaminase (AID) and proteins of the ten-eleven translocation (TET) family107. AID removes the amino group from 5mC, generating a mismatch between thymine (T) and guanine (G) bases, which is then recognized and corrected by thymine DNA glycosylase (TDG) and other DNA repair mechanisms, ultimately resulting in loss of methylation108. Proteins of the TET family (TET1, TET2 and TET3) catalyse the oxidative conversion of 5mC to 5-hydroxymethylcytosine (5hmC), which can be then either further oxidized to form 5-formylcytosine (5fC) and 5-carboxylcytosine (5caC) or deaminated to form 5-hydroxymethyl-uracil (5hmU). These intermediates are then replaced by cytosine via TDG and base excision repair mechanisms109–111. Unlike the DNMT-deficient mice, mice deficient in AID, TET1 or TET2, or both TET1 and TET2, are viable112–114, but deletion of Tet3 is neonatal lethal115, suggesting that TET3 is more important than other DNA methylation erasers in early development.
DNA methylation in AKI and repair
Although DNA methylation was the first epigenetic mark to be identified, the study of DNA methylation in kidney diseases is in its infancy. The involvement of DNA methylation changes has been implicated in chronic kidney disorders, including CKD116–118 and diabetic kidney disease119,120. By contrast, although evidence is available to support a role for DNA methylation in AKI and repair, research into this area is limited.
DNA methylation in AKI
A 2006 study reported demethylation of a cytosine in the IFNγ responsive element within the promoter region of complement factor C3 in response to cold ischaemia and warm reperfusion in rat kidneys121. A follow-up study indicated that demethylation of the C3 promoter not only occurred in the acute phase of injury but also persisted in the chronic injury phase, suggesting that the aberrant demethylation could contribute to the pathological upregulation of C3 in IRI122. In another study, hypomethylation of the promoter region of Slc22a12, which encodes the urate transporter 1 responsible for urate reabsorption, was observed in the plasma of mice with contact-freezing-induced AKI123. In humans, hypermethylation of the promoter region of KLK1, encoding renal kallikrein, was observed in blood DNA from patients with established AKI124, and hypermethylation of the promoter of CALCA, encoding calcitonin, was observed in the urine from kidney transplant recipients125. These studies indicate that the status of DNA methylation is altered in the promoter regions of specific genes in AKI (TABLE 2), suggesting the potential use of methylation status as a biomarker for AKI.
Table 2|.
DNA methylation alterations in AKI and repair
| Gene | Model | DNA methylation alteration region | DNA methylation level change | Effects on gene expression | Detection method | Refs |
|---|---|---|---|---|---|---|
| C3 | Rat IRI, kidney | The IFNγ-responsive element in C3 promoter | ↓ | NA | Bisulfite sequencing | 121 |
| C3 | Rat kidney transplantation, kidney | IL-1-IL-6 response element and NF-κB binding site 2 in C3 promoter | ↓ | NA | Pyrosequencing | 122 |
| Slc22a12 | Mouse contact-freezing-induced AKI, plasma | Promoter | ↓ | NA | TaqMan-based unmethylated DNA-specific PCR and bisulfite sequencing | 123 |
| KLK1 | Patients with AKI, blood | Promoter | ↑ | NA | Pyrosequencing | 124 |
| CALCA | Kidney transplant patients, urine | Promoter | ↑ | NA | Methylation-specific PCR | 125 |
| Cxcl10 and Ifngr2 | Mouse IRI, kidney | Promoter | ↓5hmC | ↑ | hMeDIP-qPCR | 126 |
| Irf8 | Mouse cisplatin nephrotoxicity, kidney | 5′ UTR | ↓ | ↑ | RRBS | 129 |
| Rasali | Mouse unilateral IRI, kidney | Promoter | ↑ | ↓ | DNA MeDIP | 133 |
5hmC, 5-hydroxymethylcytosine; 5′ UTR, 5′ untranslated region; AKI, acute kidney injury; hMeDIP-qPCR, hydroxymethylated DNA immunoprecipitation-quantitative PCR; IRI, ischaemia–reperfusion injury; MeDIP, methylated DNA immunoprecipitation; NA, not available; RRBS, reduced representation bisulfite sequencing.
Using dot blot and immunohistochemical analysis, Huang et al.126 showed that the global level of 5hmC was decreased following ischaemic AKI in mice whereas the global level of 5mC was unchanged. Intriguingly, the decrease in 5hmC was associated with reduced expression of TET1 and TET2, the enzymes that oxidize 5mC to 5hmC in the process of DNA demethylation, suggesting a mechanism for the reduction in 5hmC. In addition, DNA hydroxymethylome profiling data showed that lower levels of 5hmC in the promoter regions of Cxcl10 (encoding CXC-chemokine ligand 10) and Ifngr2 (encoding IFNγ receptor 2) were associated with increased expression of these two genes after IRI. Subsequent work showed that genes associated with IRI exhibited significantly higher 5hmC enrichment in their gene body regions, leading the researchers to suggest that hyper-hydroxymethylation in the gene body alters the expression of these genes in AKI127. In contrast to the findings by Huang et al.126, a more recent and more quantitative analysis of the genome-wide DNA methylation status of mouse renal tissues using reduced representation bisulfite sequencing (RRBS) found that both acute and chronic IRI (involving reperfusion for 24 h or 7 days, respectively) induced genome-wide DNA demethylation128. This demethylation status was apparent in analyses of overall DNA methylation status as well as in analyses of promoter, exon and intron regions. The discrepancy between these two studies might relate to the different degrees of injury induced and the different detection methods used. The latter study used RRBS with higher sensitivity. The researchers also identified 200 and 191 genes that had differentially methylated promoter regions in acute and chronic IRI, respectively. Of these genes, 79 were common to both conditions, and further investigation indicated that promoter methylation alterations contributed to differences in their expression.
Using RRBS, we performed genome-wide DNA methylation profiling and unveiled widespread DNA methylation alterations throughout the genome in kidney tissues from mice with cisplatin-induced AKI129. Treatment of cultured rat proximal tubular cells with the DNA methylation inhibitor 5-aza-2′-deoxycytidine augmented cisplatin-induced apoptosis, and in line with these findings, genetic ablation of Dnmt1 in proximal tubular cells exacerbated cisplatin-induced AKI in mice. Furthermore, hypomethylation and induction of interferon regulatory factor 8 (Irf8) by cisplatin treatment contributed to renal tubular cell apoptosis, demonstrating a pathological role of DNA methylation in the pathogenesis of AKI129.
Clearly, the study of DNA methylation in AKI is an emerging area and further research is needed in a number of areas. First, genome-wide DNA methylation profiling of additional mouse models other than ischaemic and cisplatin-induced AKI is needed, as well as profiling of DNA methylation status in patients with AKI. The newly developed microarray- and deep-sequencing-based technologies will facilitate the quantitative analysis of DNA methylation sites at single-nucleotide resolution or even at single-cell resolution across the genome (BOX 1). These technological advances are particularly important as the DNA methylation profiles of specific cell types represent another area that requires further study. The kidney is a highly complex organ with many different cell types. Notably, different cell types can respond differently to kidney injury. Because whole-kidney DNA methylation profiling reveals the overall DNA methylation changes of various cell types, cell-type-specific DNA methylation profiling is needed to better understand DNA methylation changes and regulation in AKI. In addition, the role of DNA methylation in AKI requires better understanding; although available limited evidence supports a role of DNA methylation in the process of AKI, the exact role is unclear. The roles of DNA methylation writers, readers and erasers in AKI also remain unknown and require further study using pharmacological and genetic modulation of DNA methylation in models of AKI. Finally, the function of the 5mC derivative, 5hmC, remains largely unclear, although it is thought to have profound effects on gene regulation130. A moderate level (0.15–0.17%) of 5hmC was observed in the kidney131; however, as mentioned above, 5hmC levels decrease after ischaemic AKI, although the relevance of this decrease is unclear126.
Box 1 | Tools for the analysis of DNA methylation.
New microarray- and deep-sequencing-based technologies will facilitate the quantitative analysis of DNA methylation sites at single-nucleotide or single-cell resolution across the genome. The Illumina Infinium Methylation BeadChip array is a microarray-based method that is cost-effective and requires a small amount of input DNA, enabling >850,000 methylation sites to be interrogated at single-nucleotide resolution with exceptional high coverage of RefSeq genes (which define genomic sequences that can be used as reference standards for well-characterized genes) and CpG islands. The drawback of this method is that it is currently applicable only to human samples. Reduced representation bisulfite sequencing (RRBS) is a deep-sequencing technique that is as sensitive as whole-genome bisulfite sequencing but is more cost-effective as it involves sequencing of a representative sample of the whole genome, covering about 60% of promoters and 85% of CpG islands. The obvious disadvantage of RRBS is that it captures most, but not all, CpG sites in the genome. Genome-wide DNA methylation analysis not only reveals novel epigenetic changes that occur in AKI but also facilitates the identification of new therapeutic targets.
DNA methylation in kidney repair
Maladaptive or incomplete repair after AKI can lead to renal fibrosis. Although DNA methylation has been explored quite extensively in the context of renal fibrosis in CKD, only a few studies have investigated the role of DNA methylation in the context of AKI progression.
DNA methylation profiling has revealed that kidney samples from patients with CKD exhibit distinct DNA methylation profiles compared with samples from healthy controls116–118. A 2017 study identified methylation levels of 19 CpG sites in whole blood associated with estimated glomerular filtration rate and CKD, 5 of which showed consistent changes in biopsy samples from patients with CKD132. Other studies have shown that pharmacological inhibition of DNA methylation (using DNA methylation inhibitors, such as 5′-azacytidine and 5-aza-2′-deoxycytidine or a DNA demethylation activator, such as hydralazine) attenuates renal fibrosis in various mouse models of CKD; similarly, heterozygous Dnmt1-knockout mice are also protected from fibrosis in CKD models13–16. One study has examined the effects of inhibiting DNA methylation on AKI progression to CKD; in that study, suppression of DNA methylation with low-dose hydralazine ameliorated renal fibrosis and prevented the progression of AKI to CKD in a model of IRI133.
Expression of RASALI, which encodes the tumour suppressor RAS GTPase-activating-like protein 1, is suppressed in fibroblasts isolated from human fibrotic kidneys and in a number of mouse models of CKD, in association with hypermethylation of its promoter region13–15. Silencing of Rasal1 by promoter hypermethylation leads to persistent activation of RAS, which contributes to the sustained activation of fibroblasts and renal fibrosis13. A 2017 study further showed that low-dose hydralazine induced TET3 expression and promoted demethylation of the Rasal1 promoter, resulting in attenuation of renal fibrosis in a model of unilateral IRI, suggesting that hypermethylation of the Rasal1 promoter is involved in maladaptive kidney repair after AKI133.
The renoprotective protein Klotho is primarily expressed in tubular cells; its expression is reduced in patients and animal models of AKI and CKD, and it is being investigated as a potential therapeutic for AKI and CKD134,135. The suppression of Klotho in renal fibrosis is now understood to be regulated by DNA methylation. Hypermethylation of the KL promoter was demonstrated in kidney tissues and peripheral blood mononuclear cells from patients with CKD136 as well as in the animal models of UUO, ciclosporin-A-induced nephropathy and adenine-induced CKD16,137–139. Use of the DNA methylation inhibitor 5-aza-2′-deoxycytidine or knockdown of DNMT1 or DNMT3A inhibited hypermethylation of the KL promoter and preserved Klotho expression16,137,139. Although no direct evidence has shown that DNA methylation regulation of KL is involved in the progression of AKI to CKD, the above evidence suggests that this is a possibility.
Non-coding RNAs
Less than 2% of the human genome is transcribed into RNAs with protein-coding potential; the majority is transcribed into non-coding RNAs. Non-coding RNAs are classified according to their size as either small non-coding RNAs (<200 nucleotides) or long non-coding RNAs (lncRNAs) (>200 nucleotides). These RNAs cannot be translated into proteins but act as crucial epigenetic modulators of gene expression.
Of the endogenous small non-coding RNAs, microRNAs have been most extensively investigated. MicroRNAs usually contain 21–25 nucleotides and act as post-transcriptional inhibitory regulators of gene expression (FIG. 4a), predominantly by binding to the 3′ untranslated region of their target gene mRNAs, which either induces degradation of the mRNA or, more commonly, blocks translation of the mRNA into protein140,141. According to miRBase 22, approximately 2,600 mature microRNAs have been identified in humans, which are thought to regulate more than half of the human genome.
Fig. 4 |. Mechanisms of gene regulation by non-coding RNAs.
a | MicroRNA biogenesis requires initial transcription of a microRNA gene by RNA polymerase II into a large primary transcript that contains multiple hairpin loop structures, known as a primary microRNA (pri-microRNA). This transcript is processed by the ribonuclease, Drosha, exported from the nucleus and processed by another enzyme, called Dicer, to produce a microRNA–microRNA duplex in the cytoplasm consisting of a guide strand and a passenger strand. This complex is unwound and the mature microRNA (the guide strand) is incorporated into the ribonucleoprotein complex, RISC (RNA-induced silencing complex), which is guided to target genes, leading to post-transcriptional silencing either by mRNA degradation or most commonly by translational repression. b | Long non-coding RNAs (lncRNAs) can regulate gene expression at both transcriptional (1–4) and post-transcriptional levels (5–7). At a transcriptional level, lncRNAs can (1) recruit transcription activators or repressors to the target gene, resulting in gene activation or suppression, respectively; (2) act as decoy factors by binding and sequestering transcription factors and other proteins or regulatory RNAs away from chromatin, thereby inhibiting gene transcription; (3) guide chromatin-modifying enzymes to specific gene targets and then modulate chromatin states either in cis or trans, leading to positive or negative regulation of gene expression; and (4) serve as a structural scaffold to recruit multiple proteins or RNAs to form transcription regulatory complexes, which can positively or negatively regulate gene expression. lncRNAs can also regulate gene expression at a post-transcriptional level by (5) binding to splicing factors to regulate RNA splicing, (6) directing RNA-binding proteins to the target mRNAs to either promote or inhibit protein translation or (7) serving as a molecular sponge that binds and titrates microRNAs away from their targets.
lncRNAs exist in both the cell nucleus and cytoplasm. Unlike microRNAs, which regulate gene expression exclusively at a post-transcriptional level, lncRNAs can function both transcriptionally and post-transcriptionally by interacting with DNA, RNA and protein (FIG. 4b). lncRNAs can be categorized according to their genomic location as sense, antisense, bidirectional, intronic, intergenic, enhancer or circular lncRNAs. They can also be classified according to their molecular function at the transcriptional level as signal lncRNAs, which act as molecular signals of transcription activity in spatial-dependent and temporal-dependent manners; decoy lncRNAs, which inhibit transcription by binding and sequestering transcription factors and other proteins or regulatory RNAs away from chromatin; guide lncRNAs, which modulate chromatin states and gene expression by guiding chromatin-modifying enzymes to specific gene targets; and scaffold lncRNAs, which facilitate the assembly of transcription regulatory complexes that positively or negatively regulate gene expression142. In addition, lncRNAs can participate in post-transcriptional regulation. For example, lncRNAs can affect mRNA processing, translocation and stability by binding to the complementary RNAs and can also interact with splicing factors to modulate RNA splicing. lncRNAs can also recruit RNA-binding proteins to target mRNAs to promote or inhibit protein translation. Moreover, they can bind to microRNAs to titrate them away from their targets. Furthermore, some lncRNAs act as precursors for small regulatory RNAs that can be processed into small interfering RNAs, microRNAs or piwi-interacting RNAs, with effects on gene expression143,144. To date, approximately 30,000 lncRNAs have been identified in the human genome; the functions of about 300 of these are annotated in the Long Non-coding RNA database v2.0 (REF.145).
Non-coding RNAs in AKI and kidney repair
Non-coding RNAs, particularly microRNAs and lncRNAs, have emerged as important epigenetic regulators of AKI and kidney repair. In addition, circulating non-coding RNAs might represent potential biomarkers of AKI.
MicroRNAs in AKI
By regulating gene expression, microRNAs regulate diverse cellular and physiological processes, such as proliferation, differentiation, cell death and organ development146–149. MicroRNAs have also been implicated in the pathogenesis of various human diseases149–153.
An accumulating body of evidence suggests that microRNAs are critical regulators of renal development and pathophysiology154–160. Given that the role of microRNAs in kidney diseases has been extensively reviewed else-where156,157,161–167, we focus here on the recent research progress of microRNA in AKI and repair.
MicroRNAs in ischaemic AKI.
We demonstrated a role for microRNAs in AKI by generating conditional Dicer1 (which encodes Dicer)-knockout mice, in which over 80% of microRNAs were depleted specifically in kidney proximal tubules168. These mice are remarkably resistant to ischaemic AKI, suggesting a pathogenic role of microRNAs in ischaemic AKI.
Microarray analyses by our group and others have demonstrated that the expression levels of microRNAs markedly alter in response to ischaemic AKI in mice168–171. Furthermore, a number of microRNAs have been identified to have a pathogenic or protective role in ischaemic AKI (TABLE 3). Some of these microRNAs, including miR-24 (REF. 172), miR-150 (REF. 173), miR-494 (REF. 174) and miR-687 (REF. 175), are pathogenic and contribute to kidney injury, whereas others, including miR-17–5p176, miR-26a177, miR-126 (REF. 178), miR-127 (REF. 179), miR-146a169,180,181, miR-205 (REF. 182) and miR-489 (REF. 183) protect against ischaemic AKI. miR-21 has in general been shown to be protective in ischaemic AKI169,184–187; however, one study showed that overexpression of miR-21 increased apoptosis of cultured rat kidney epithelial cells in response to hypoxia–reoxygenation and that inhibition of miR-21 preserved expression of the intracellular membrane trafficking protein Rab11a and ameliorated autophagy activation, protecting against renal IRI in rats188.
Table 3|.
MictoRNAs in AKI
| MictoRNA | Upstream effector | Target | Function | Model | Response | Manipulation | Refs |
|---|---|---|---|---|---|---|---|
| MicroRNAs in ischaemic AKI | |||||||
| miR-24 | NA | S1P1, H2AX and HO1 | Pathogenic | Kidney transplantation patients, mouse IRI, HK-2 cell ATP depletion and endothelial cell hypoxia–reoxygenation | ↑ | • In vitro: mimic and anti-miR •In vivo: anti-miR |
172 |
| miR-150 | NA | IGF1Ra | Pathogenic | Mouse IRI | ↓ | miR-150-deficient mice | 173 |
| miR-494 | NA | ATF3 | Pathogenic | Mouse IRI and urine from patients with AKI | ↑ | In vivo: mimic | 174 |
| miR-687 | HIF1 | PTEN | Pathogenic | Mouse IRI, HEK cells and BUMPT cell hypoxia | ↑ | • In vitro: mimic and anti-miR • In vivo: anti-miR |
175 |
| miR-17–5p | p53 | DR6 | Protective | Mouse IRI and RPTC hypoxia | ↑ | • In vitro: mimic and anti-miR • In vivo: mimic |
176 |
| miR-21 | HIF1α | PDCD4, TSP1 and Rab11a | Protective and/or pathogenic | Mouse IPC, mouse IRI, rat IRI, urine and plasma from patients with AKI, NRK-52E cell hypoxia–reoxygenation, cultured human renal epithelial cell CoCl2 or hypoxia | ↑ | • In vitro: mimic and anti-miR • In vivo: anti-miR |
169,184–188 |
| miR-26a | NA | IL-6 | Protective | Mouse IRI | ↓ | In vivo: mimic | 177 |
| miR-126 | NA | PI3KR2 | Protective | Mouse IRI | NA | miR-126 conditional overexpressing mice | 178 |
| miR-127 | HIF1α | KIF3B | Protective | Rat IRI, and NRK-52E cell and HK-2 cell hypoxia–reoxygenation | ↑ | In vitro: mimic and anti-miR | 179 |
| miR-146a | NF-κB | IRAK1 | Protective | Kidney transplantation patients, mouse unilateral IRI and LPS-challenged kidney following IRI | ↑ | • In vitro: mimic and anti-miR • In vivo: anti-miR and miR146a-deficient mice |
169,180,181 |
| miR-205 | NA | EGLN2 | Protective | HK-2 cell hypoxia–reoxygenation | ↓ | In vitro: mimic and anti-miR | 182 |
| miR-489 | HIF1α | PARP1 | Protective | Mouse IRI, RPTC hypoxia and ATP depletion | ↑ | • In vitro: mimic and anti-miR • In vivo: anti-miR |
183 |
| MicroRNAs in cisplatin nephrotoxicity | |||||||
| miR-34a | p53 | SIRT1a | Protective | Mouse, BUMPT cells, NRK-52E cells | ↑ | In vitro: mimic and anti-miR | 191,192 |
| miR-125b | NRF2 | AhRR | Protective | Mouse, NRK-52E cells | ↑ | In vitro: mimic | 193 |
| miR-155 | NA | c-Fos | Protective | Mouse | NA | miR-155-deficient mice | 195 |
| miR-122 | NA | FOXO3 | Pathogenic | Mouse, NRK-52E cells | ↓ | In vitro: mimic and anti-miR | 192 |
| miR-146b | NA | ErbB4 | Pathogenic | Serum from patients with AKI, rat, NRK-52E cells | ↑ | In vitro: anti-miR | 194 |
| miR-181a | NA | BCL-2 | Pathogenic | HK-2 cells | ↑ | In vitro: anti-miR | 196 |
| miR-375 | p53 and NF-kB | HNF1β | Pathogenic | Mouse and RPTCs | ↑ | In vitro: anti-miR | 198 |
| miR-449 | NA | SIRT1a | Pathogenic | NRK-52E cells | ↑ | In vitro: anti-miR | 197 |
| miR-709 | NA | mtTFA | Pathogenic | Kidney biopsy samples from patients with AKI, mouse and mPTCs | ↑ | • In vitro: mimic and anti-miR • In vivo: anti-miR |
199 |
| MicroRNAs in other types of AKI | |||||||
| miR-150 | NA | IGF1Ra | Pathogenic | Myocardial-infarction-induced AKI | NA | miR-150-deficient mice | 173 |
| miR-192 | NA | MDM2 | Pathogenic | Aristolochic acid nephropathy | ↑ | NA | 200 |
AhRR, aryl hydrocarbon receptor repressor; AKI, acute kidney injury; anti-miR, anti-microRNA oligonucleotides; ATF3, activating transcription factor 3; BCL-2, B cell lymphoma 2; BUMPT cells, a mouse renal proximal tubular cell line; DR6, death receptor 6; EGLN2, Egl nine homologue 2; ErbB4, erb-B2 receptor tyrosine kinase 4; HEK cells, human embryonic kidney cells; HIF1, hypoxia-inducible factor 1; HK-2 cells, an immortalized human proximal tubular epithelial cell line; HNF1β, hepatocyte nuclear factor 1 homeobox B; HO1, haem oxygenase 1; IGF1R, insulin-like growth factor 1 receptor; IPC, ischaemic preconditioning; IRAK1, IL-1 receptor-associated kinase 1; IRI, ischaemia-reperfusion injury; KIF3B, kinesin family member 3B; LPS, lipopolysaccharide; MDM2, murine double-minute 2; mPTCs, mouse proximal tubular epithelial cells; mtTFA, mitochondrial transcriptional factor A; NA, not available; NRF2, nuclear factor erythroid-2-related factor 2; NRK-52E cells, a rat renal proximal tubular cell line; PARP1, poly(ADP-ribose) polymerase 1; PDCD4, programmed cell death protein 4; PI3KR2, phosphoinositide 3-kinase regulatory subunit 2; PTEN, phosphatase and tensin homologue; RPTC, rat proximal tubular cell; S1P1, sphingosine-1-phosphate receptor 1; SIRT1, sirtuin 1; TSP1, thrombospondin 1.
An indirect target.
Our group has elucidated the roles of three specific microRNAs in ischaemic AKI, including miR-687, miR-489 and miR-17–5p175,176,183. All of these microRNAs are markedly induced in both in vivo and in vitro models of ischaemic AKI. The induction of miR-687 and miR-489 is mediated by hypoxia-inducible factor 1 (HIF1). Induction of miR-678 suppresses phosphatase and tensin homologue (PTEN) — a known inducer of G1 arrest — which in turn promotes cell cycle progression and apoptosis and exacerbates ischaemic AKI175. By contrast, miR-489 and miR-17–5p have a renoprotective role in ischaemic AKI. Overexpression of miR-489 in an in vitro model of ischaemic AKI attenuated cell apoptosis, whereas inhibition of miR-489 increased apoptosis in vitro and aggravated ischaemic AKI in mice. Moreover, PARP1 — a known cell death mediator in ischaemic AKI — was identified as a target gene of miR-489, suggesting that miR-489 protects against ischaemic AKI by repressing PARP1 (REF. 183). Upregulation of miR-17–5p was induced by p53, which led to suppression of death receptor 6 (DR6; also known as TNFRSF21), resulting in inhibition of apoptosis in vitro and renoprotective effects in vivo176.
MicroRNAs are also involved in the regulation of inflammation in ischaemic AKI. For instance, miR-146a regulates inflammation via its inhibitory effects on the Toll-like receptor (TLR)-NF-κB pathway189,190. Several studies have reported increased expression of miR-146a in mice with renal IRI and suggested that it has a renoprotective role in ischaemic AKI169,180,181. Deficiency of miR-146a aggravates tubular injury, increases inflammation and increases fibrosis in mice subjected to unilateral IRI181. Moreover, knockdown of miR-146a neutralized the protective effect of LPS pretreatment on renal IRI180. Mechanistically, activation of the TLR-NF-κB pathway after renal IRI initiates an acute inflammatory response but also upregulates miR-146a, which targets IL-1 receptor-associated kinase 1 (IRAKI), the upstream target of NF-κB, which in turn negatively regulates TLR-NF-κB signalling and suppresses inflammation. By contrast, miR-26a is downregulated in response to IRI in mice177. Overexpression of miR-26a by tail-vein injection of a recombinant lentiviral vector containing the miR-26a precursor, pre-mir-26a, protected against ischaemic AKI and promoted expansion of FOXP3+CD4+ regulatory T cells by repressing IL-6. Moreover, administration of miR-26a had renoprotective effects even when given after IRI, suggesting that this approach might have therapeutic potential.
Another microRNA, miR-126, regulates vascular regeneration by mobilizing haematopoietic stem and progenitor cells. miR-126 targets PI3KR2 (phosphoinositide 3-kinase regulatory subunit 2; also known as PI3-kinase subunit p85β), a key regulator of the PI3K–AKT (protein kinase B) signalling pathway, and affects the expression of genes that are essential for angiogenesis and inflammation. In ischaemic AKI, mice with overexpression of miR-126 in the haematopoietic compartment had better renal function and lower levels of tubular damage, inflammation and fibrosis than control mice178. The beneficial effects of miR-126 were attributed to the preservation of microvascular integrity by enhancing vasculogenic progenitor cell mobilization after ischaemic AKI178.
MicroRNAs in cisplatin-induced AKI and other forms of AKI.
A number of microRNAs have also been implicated in the pathogenesis of cisplatin-induced AKI163,191–199 (TABLE 3). Our laboratory examined cisplatin-induced AKI in mice with proximal-tubule-specific deletion of Dicer1, which, to our surprise, developed a similar level of AKI in response to cisplatin treatment as wild-type mice, implying that overall depletion of microRNAs does not significantly affect cisplatin-induced AKI198. We also assessed changes in the expression of microRNAs using microarray analysis, leading to the identification of miR-375 as a pathogenic microRNA. This microRNA was significantly induced in response to NF-κB and p53 following cisplatin treatment, which in turn suppressed expression of the anti-apoptotic factor HNF1β (hepatocyte nuclear factor 1 homeobox B) and thereby induced renal tubular cell death. This study therefore uncovered a novel p53-NF-κB-miR-375-HNF1β signalling pathway that contributes to cisplatin-induced nephrotoxicity during chemotherapy. Another study showed that induction of miR-449 after cisplatin treatment in cultured renal tubular epithelial cells induced inhibition of SIRT1, increased acetylation of p53 and increased expression of the pro-apoptotic factor BAX (BCL-2 (B cell lymphoma 2)-associated X protein)197. Accordingly, blockade of miR-449 suppressed cell apoptosis induced by cisplatin in vitro via preservation of the SIRT1–p53–BAX signalling pathway. Moreover, miR-146b was found to be upregulated in cisplatin-treated renal tubular epithelial cells and cisplatin-treated rats194. The researchers further showed that miR-146b directly targets a regulator of cell proliferation and differentiation — erb-B2 receptor tyrosine kinase 4 (ErbB4). Suppression of miR-146b attenuated cisplatin nephrotoxicity in cultured tubular epithelial cells by inducing expression of ErbB4 and increasing cell proliferation. Finally, a 2018 study showed that miR-709, which is induced in mouse and human kidney samples following cisplatin treatment, targets the gene encoding mitochondrial transcriptional factor A (mtTFA) to induce mitochondrial dysfunction, thereby contributing to cisplatin-induced AKI199.
In addition to ischaemic and cisplatin-induced AKI, microRNAs have been implicated in other types of AKI (TABLE 3). For example, miR-150 has been linked to myocardial-infarction-induced AKI; mice deficient in miR-150 were protected from inflammation, interstitial cell apoptosis and AKI following myocardial infarction173. In a mouse model of aristolochic-acid-induced nephropathy, induction of miR-192 in proximal tubular epithelial cells suppressed expression of MDM2 (murine double-minute 2), leading to activation of p53 and G2/M cell cycle arrest200.
MicroRNAs as biomarkers for AKI
MicroRNAs are remarkably stable and can be detected and quantified in various body fluids, including plasma and urine201. The altered expression profile of specific microRNAs in AKI has led several groups to suggest that they could be used as biomarkers for the diagnosis of AKI202–212 (TABLE 4). For example, miR-21 has been consistently reported to be highly elevated in urine and plasma samples of patients with AKI as well as in animal models of IRI204–206. A 2011 study identified and validated the downregulation of miR-16 and miR-320 and upregulation of miR-210 in the plasma of patients with AKI (n = 77) compared with levels in healthy controls (n = 30)202. Another urinary microRNA profiling study revealed four microRNAs (miR-21, miR-200c, miR-423 and miR-4640) that were consistently changed in patients with AKI206, whereas another identified a novel set of microRNAs, including miR-101-1-3p, miR-127-3p, miR-210-3p, miR-126-3p, miR-26b-5p, miR-29a-3p, miR-146a-5p, miR-27a-3p, miR-93-3p and miR-10a-5p, as downregulated in the serum of patients with AKI in an intensive care unit (n = 35) compared with levels in healthy controls (n = 20)211. This study also found that miR-26b-5p, miR-146a-5p, miR-93-3p and miR-127-3p had value as biomarkers to predict AKI predisposition in patients undergoing cardiac surgery as their levels were downregulated days before serum creatinine increased211.
Table 4|.
potential microRNA biomarkers for the diagnosis of AKI
| MictoRNA | Model | Source | Species | Response | Refs |
|---|---|---|---|---|---|
| miR-210, miR-16 and miR-320 | Patients with AKI | Plasma | Human | ↑ (miR-16 and miR-320↓) | 202 |
| miR-21 and miR-155 | Patients with AKI | Urine | Human | ↑ (miR-155↓) | 204 |
| miR-21 | Patients with AKI after cardiac surgery | Plasma and urine | Human | ↑ | 205 |
| miR-494 | Patients with AKI | Urine | Human | ↑ | 174 |
| miR-21, miR-200c, miR-423 and miR-4640 | Patients with AKI | Urine | Human | ↑ (miR-4640↓) | 206 |
| miR-101–1-3p, miR-127–3p, miR-210–3p, miR-126–3p, miR-26b-5p, miR-29a-3p, miR-146a-5p, miR-27a-3p, miR-93–3p and miR-10a-5p | Patients with AKI | Serum | Human | ↓ | 211 |
| miR-10a and miR-30d | Ischaemic AKI | Urine | Mouse | ↑ | 203 |
| miR-10a, miR-192 and miR-194 | Ischaemic AKI | Plasma | Rat | ↑ | 208 |
| miR-714, miR-1188, miR-1897–3p, miR-877 and miR-1224 | Ischaemic AKI | Plasma and kidney | Mouse | ↑ | 210 |
| let-7g-5p, miR-93–5p, miR-191a-5p and miR-192–5p | Cisplatin nephrotoxicity | Urine | Rat | ↑ | 207 |
| miR-200a–c, miR-192 and miR-194 | Contact-freezing-induced AKI | Plasma | Mouse | ↑ | 212 |
AKI, acute kidney injury
All the aforementioned studies demonstrate the potential of microRNAs as biomarkers for AKI; however, their specificity for AKI requires consideration. Some microRNAs are associated not only with AKI but also with other diseases, including other kidney diseases. For example, miR-21 is increased in AKI; however, serum miR-21 level is also significantly increased in patients with stroke or atherosclerosis213, and urinary miR-21 levels are associated with CKD214. Moreover, microRNAs in different body fluids and blood fractions may have different biological relevance. For example, plasma levels of miR-210 were described by one study as an independent prognostic indicator of survival in AKI202, but serum miR-210 levels were shown by another to be decreased in patients with AKI211. The involvement of many different microRNAs in AKI indicates that a panel of microRNAs would potentially be more useful than a single microRNA as a biomarker. Further research is needed in this area.
MicroRNAs in kidney repair
Despite intensive investigation of the role of microRNAs in AKI, a paucity of information exists regarding the role of microRNAs in kidney repair following AKI. A 2016 study used small RNA sequencing (RNA-seq) to generate temporal microRNA expression profiles, and 103 microRNAs were found to be differentially expressed during the injury and repair phase in a model of folic-acid-induced AKI171. Among these, three microRNAs were highly abundant and differentially expressed at different phases of injury: miR-18a-5p was induced during the acute injury phase, miR-132–3p was upregulated during the repair phase and miR-146b-5p was upregulated in the fibrotic phase. However, the function of these microRNAs was not investigated. Studies from the past few years have, however, indicated that some microRNAs that are induced during injury are possibly involved in kidney repair after AKI. For instance, induction of miR-687 during renal IRI inhibits PTEN to promote cell proliferation — an effect that might facilitate tissue repair or regeneration in ischaemic AKI175. On the other hand, miR-423–5p suppressed GSTM1 (glutathione-S-transferase Mu1) and inhibited cell proliferation, thereby preventing tissue repair following ischaemic AKI215. Similarly, miR-146b and miR-1247 limit kidney repair by inhibiting cell proliferation in cisplatin-induced or alcohol-induced AKI, respectively194,216.
lncRNAs in AKI
Although less studied than microRNAs, some evidence exists to suggest a role for lncRNAs in AKI.
Using RNA-seq to perform whole-transcriptome profiling, one study identified a set of differentially expressed lncRNAs in renal proximal tubular epithelial cells under hypoxic and inflammatory conditions217. Among these, three lncRNAs (MIR210HG, long intergenic non-coding (linc)-ATP13A4–8 and linc-KIAA1737–2) were of particular note as they were most markedly induced by hypoxia and cytokines and were detectable in human kidney biopsy samples from kidney transplant patients, but the role of these lncRNAs in AKI and repair is unclear. Another study used RNA-seq and chromatin immunoprecipitation (ChIP) sequencing of HIF1 binding sites to identify a novel, hypoxia-inducible, HIF1-dependent lncRNA called DARS antisense RNA1 (DARS-AS1)218. Silencing of DARS-AS1 stimulated apoptosis of cultured renal tubular cells in response to hypoxia, indicating that DARS-AS1 protects against hypoxia-induced cell injury. Moreover, by quantitative-PCR-based disease-related lncRNA array, lncRNA-PRINS (psoriasis associated non-protein-coding RNA induced by stress) was identified as another hypoxia-induced, HIF1α-dependent lncRNA219. lncRNA-PRINS interacts with RANTES (regulated on activation, normal T cell expressed and secreted) — an important inflammatory mediator in ischaemic AKI — to regulate its production under hypoxic conditions, suggesting a role for lncRNA-PRINS in the pathogenesis of AKI219.
Researchers have demonstrated a role for several disease-related lncRNAs, such as MALAT1 (metastasis-associated lung adenocarcinoma transcript 1; also known as NEAT2 (nuclear paraspeckle assembly transcript 2)) and GAS5 (growth arrest-specific 5), in ischaemic AKI. MALAT1, a large (6.5 kb), nuclear-enriched and highly conserved lncRNA, was one of the first lncRNAs to be associated with human disease; growing evidence indicates that MALAT1 participates in the development and progression of various cancers, including renal cell carcinoma. A 2015 study identified MALAT1 as the most strongly upregulated lncRNA in the kidneys — particularly proximal tubular cells — of mice subjected to inspiratory hypoxia220. A more recent study showed MALAT1 levels to be increased in biopsy samples from humans with ischaemic injury, plasma samples from patients with AKI, in kidneys of mice with IRI and in kidney cells cultured under hypoxia–reoxygenation conditions221. However, inhibition of MALAT1 by antisense oligonucleotides did not affect cell cycle progression, cell proliferation or apoptosis during hypoxia-reoxygenation injury in HK-2 (human kidney-2) cells. Moreover, genetic ablation of MALAT1 in mice did not affect kidney dysfunction or survival of mice following bilateral IRI or outer medullary injury, cell proliferation, interstitial fibrosis, inflammation or capillary rarefaction following unilateral IRI, indicating that MALAT1 is dispensable in the development of renal IRI and subsequent kidney repair221. A 2018 study identified GAS5 — a tumour suppressor lncRNA that regulates apoptosis and growth arrest — as a functional lncRNA in ischaemic AKI222. Similar to some of the other lncRNAs that have been linked to AKI, GAS5 is also hypoxia responsive, and its expression is increased in the kidneys of mice with renal IRI and in renal tubular epithelial cells that have undergone hypoxia–reoxygenation treatment. Knockdown of GAS5 reduced tubular cell death induced by hypoxia–reoxygenation treatment, probably by regulating expression of thrombospondin 1 (REF.222).
In contrast to findings from MALAT1-knockdown studies in ischaemic AKI, knockdown of MALAT1 inhibited LPS-induced cell death in cultured renal tubular epithelial cells223. The detrimental effects of MALAT1 in LPS-induced tubular cell death might be mediated by inhibiting the suppressive effects of miR-146a on NF-κB signalling223. Several other lncRNAs are also upregulated in septic AKI and might contribute to disease pathogenesis. Similar to MALAT1, the lncRNA PVT1 (plasmacytoma variant translocation 1) is associated with diverse cancers. Expression of PVT1 is significantly upregulated by LPS treatment and might aggravate septic AKI by modulating TNF and JNK-NF-κB signalling pathways224. Similarly, the lncRNA NEAT1 is upregulated in patients with sepsis-induced AKI, acts to suppress miR-204 and activates NF-κB signalling, thereby promoting LPS-induced cell injury225. In addition, lncRNA MEG3 (maternally expressed gene 3) exacerbated LPS-induced kidney injury by modulating the miR-21-PDCD4 (programmed cell death protein 4) axis226. Moreover, inhibition of lncRNA HOTAIR (HOX transcript antisense RNA) attenuated sepsis-induced AKI in in vitro and in vivo models by regulating the miR-22–HMGB1 (high mobility group box 1) pathway227. These data indicate that lncRNAs might serve as novel therapeutic targets for ischaemic and septic AKI; however, information relating to the role of lncRNAs in other forms of AKI are lacking.
Epigenetic writers
Specific enzymes that add epigenetic marks on histone proteins or DNA.
Epigenetic readers
Effector proteins that recognize and bind epigenetic marks.
Epigenetic erasers
Specific enzymes that remove the epigenetic marks on histone proteins or DNA.
Core histones
Histones H2A, H2B, H3 and H4, which form the nucleosome core (also known as the histone octamer).
Linker histones
Histones that bind to internucleosomal DNA (also known as linker DNA), facilitating the formation of a compact chromatin structure.
Histone acetylation
Histone modification that involves the addition of an acetyl group to the ε-amine of lysine on all four core histones by histone acetyltransferases.
Histone methylation
Histone modification that involves the addition of a methyl group to a basic amino acid on core histones by histone methyltransferases
Histone crotonylation
Histone modification that adds a crotonyl group to lysine residues on the core histones by histone crotonyltransferases.
Matrix chromatin immunoprecipitation
High-throughput chromatin immunoprecipitation method in which antibodies are immobilized in a 96-well plate and all the procedures are done on the same plate without sample transfer.
Permissive histone marks
Histone modifications that promote gene transcription.
Elongation marks
Histone modifications that promote transcription elongation.
Repressive histone marks
Histone modifications that inhibit gene transcription.
CpG dinucleotides
Regions of DNA in which a cytosine nucleotide is followed by a guanine nucleotide, connected by a phosphodiester bond.
CpG islands
Regions of DNA >200 base pairs in length that have a CG content >50% and observed CpG (number of CpGs observed in a window):expected CpG (number of Cs × number of Gs/window length) ratio ≥0.6.
Genomic integrity
Integrity of the genome or genome stability.
X chromosome inactivation
Process that inactivates one of the two X chromosomes in female mammals.
Genomic imprinting
Biological process that epigenetically marks a gene, leading to gene expression in a parent-of-origin manner.
Hemimethylated DNA
DNA that has one strand methylated and another unmethylated.
tRNAs
Class of small RNA that carries a particular amino acid to the ribosome on the basis of the mRNA nucleotide sequences.
DNA hydroxymethylome profiling
Genome-wide analysis of DNA hydroxymethylation.
Hyper-hydroxymethylation
Increased DNA hydroxymethylation.
Reduced representation bisulfite sequencing
(RRBS). Genome-wide DNA methylation analysis method based on bisulfite sequencing that involves sequencing of a reduced, representative sample of the whole genome.
Small interfering RNAs
Class of small non-coding RNAs that bind to complementary mRNAs, leading to mRNA degradation and inhibition of protein translation.
Circulating non-coding RNAs
Non-coding RNAs that are present in the body fluid.
Dicer
Double-stranded RNA endoribonuclease that cleaves long or hairpin double-stranded RNA into small interfering RNA or precursor microRNA into microRNA.
Chromatin immunoprecipitation (ChIP)
Sequencing Method combining chromatin immunoprecipitation and nextgeneration sequencing that analyses the genome- wide DNA binding sites for transcription factors or other chromatin- associated proteins.
Inspiratory hypoxia
Hypoxia condition that animals are subjected to with a low inspiratory oxygen concentration (such as 8% of oxygen).
Circulating lncRNAs might also represent biomarkers for the diagnosis and prognosis of various diseases, including AKI. Several circulating lncRNAs — including MALAT1, NEAT1 and transcript predicting survival in AKI (TapSAKI; also known as MGAT3-AS1) — are upregulated in plasma and kidney biopsy samples of patients with AKI221,225,228. Importantly, levels of NEAT1 and TapSAKI are also associated with severity of AKI225,228.
lncRNAs in kidney repair
Direct evidence linking lncRNAs to kidney repair following AKI is lacking; however, studies of lncRNAs in renal fibrosis provide some useful insights into the potential role of lncRNAs in kidney repair. One study used RNA-seq to identify a number of differentially expressed lncRNAs during the progression of renal fibrosis in mice with UUO229. Among these, 3110045C21Rik was found to regulate several fibrosis-related genes independent of TGFβ or IL-1β signalling pathways. Another two studies performed whole-transcriptome analyses in wild-type and Smad3-knockout mice with UUO, and in rats with UUO, to identify 151 SMAD3-dependent lncRNAs and 19 lncRNAs with SMAD3 binding motifs, respectively230,231. Further investigation revealed that TCONS_00088786, TCONS_01496394 and np_5318 (ErbB4-IR) contribute to TGFβ-SMAD3-mediated renal fibrosis in these models231,232. These findings suggest that lncRNAs might participate in the process of maladaptive repair after AKI, a proposal that requires further investigation.
Potential therapeutic applications
A number of epigenetic-modifying drugs, such as histone modifiers and DNA methylation inhibitors, are currently available and have been tested in preclinical models of AKI and CKD. Increasing histone acetylation with the use of HDACis typically protects kidneys from AKI and promotes kidney repair. Similarly, inhibition of DNA methylation with the use of DNA methylation inhibitors (such as 5′-azacytidine and 5-aza-2′-deoxycytidine) or DNA demethylation activators (such as hydralazine) ameliorates renal fibrosis. Findings from preclinical studies suggest that targeting epigenetic mechanisms is a promising approach for the treatment of AKI and associated kidney diseases. Encouragingly, some of the epigenetic drugs are already in clinical use for other diseases, including the HDACis SAHA (vorinostat) for cutaneous T cell lymphoma and VPA for seizures and bipolar disorder, the DNA methylation inhibitors 5′-azacytidine (azacitidine) for myelodysplastic syndrome and 5-aza-2′-deoxycytidine (decitabine) for myelodysplastic syndrome and acute myeloid leukaemia and the DNA demethylation activator hydralazine for hypertension233. Of note, some of these drugs might be nephrotoxic, but none have been used in clinical trials for kidney diseases233. Preclinical studies have shown that some epigenetic drugs, such as HDACis, exert renoprotective effects at low doses but are nephrotoxic at higher doses46,48. Another concern about epigenetic drugs is that they are highly unspecific and induce global epigenetic changes that are not gene-specific or organ-specific. A 2017 description of a novel high-fidelity CRISPR–Cas9-based method to enable hydroxymethylation of specific genes is therefore of interest. Fusion of the catalytic domain of the DNA methylation eraser TET3 to an endonuclease deactivated high-fidelity Cas9 (dHFCas9) created a construct with which to demethylate genes that were specifically targeted by guiding RNAs17. Use of this method led to the successful reactivation of Rasal1 and Kl (two anti-fibrotic genes that are hypermethylated and silenced in renal fibrosis), resulting in the attenuation of UUO-associated renal fibrosis17. This new technology could open up new avenues for potential therapeutic applications. Moreover, microRNA-based therapy has gained great interest in AKI and kidney repair, with the identification of a number of microRNAs linked to AKI pathogenesis (TABLE 3). Manipulation of specific microRNAs with the use of microRNA mimics or anti-microRNA oligonucleotides might attenuate AKI and/or promote kidney repair and recovery. However, no microRNAs are in clinical trials for AKI right now. A few microRNAs to date are already in clinical trials of other diseases234. In particular, an anti-miR-21 (RG-012) is now in a phase II clinical trial for Alport syndrome, a genetic disorder associated with kidney dysfunction.
Conclusions
AKI is now recognized as one of the most deadly diseases worldwide; however, the available treatment is mainly supportive. The past few years have witnessed rapid progress in our understanding of the contribution of epigenetic mechanisms to kidney function and disease, including AKI and associated pathologies, shedding light on potential new diagnostic and therapeutic strategies. These studies have also highlighted the complexity of epigenetic modifications and the network of interacting factors involved in processes such as histone modifications and DNA methylation, and in gene regulation by non-coding RNAs, providing better understanding of the fundamental elements of epigenetic regulation, specifically in AKI. Further research in this area will continue to identify new therapeutic targets as well as sensitive and specific diagnostic biomarkers for AKI and its progression to CKD.
Key points.
Acute kidney injury (AKI) and subsequent kidney repair are associated with substantial epigenetic changes that have important roles in the pathogenesis and outcome of AKI.
An overall increase in histone acetylation (for example, with the use of histone deacetylase inhibitors) might attenuate AKI and promote kidney repair, but the enzymes and downstream genes that mediate these effects remain elusive.
DNA methylation might also affect AKI and kidney repair via modulation of downstream genes, but the nature of this regulation remains largely unknown.
MicroRNAs are important factors in the regulation of AKI and kidney repair, but they can be pathogenic or protective depending on the specific microRNA species.
Additional research into the epigenetic mechanisms underlying AKI may lead to the discovery of novel biomarkers and therapies for AKI.
Acknowledgements
The authors are supported in part by grants from the US National Institutes of Health (DK058831 and DK087843) and the US Department of Veterans Affairs (BX000319). Z.D. is a recipient of the Senior Research Career Scientist award from the Department of Veterans Affairs.
Footnotes
Competing interests
The authors declare no competing interests.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Reviewer information
Nature Reviews Nephrology thanks J. Lorenzen, A. B. Sanz, T. Tanaka and S. Zhuang for their contribution to the peer review of this work.
RELATED LiNKs
Long Noncoding RNA Database v2.0: http://www.lncrnadb.org miRBase 22: http://www.miRbase.org
References
- 1.Lameire NH et al. Acute kidney injury: an increasing global concern. Lancet 382, 170–179 (2013). [DOI] [PubMed] [Google Scholar]
- 2.Mehta RL et al. Recognition and management of acute kidney injury in the International Society of Nephrology 0by25 Global Snapshot: a multinational cross-sectional study. Lancet 387, 2017–2025 (2016). [DOI] [PubMed] [Google Scholar]
- 3.Mehta RL et al. International Society of Nephrology’s 0by25 initiative for acute kidney injury (zero preventable deaths by 2025): a human rights case for nephrology. Lancet 385, 2616–2643 (2015). [DOI] [PubMed] [Google Scholar]
- 4.Linkermann A. et al. Regulated cell death in AKI. J. Am. Soc. Nephrol 25, 2689–2701 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Agarwal A. et al. Cellular and molecular mechanisms of AKI. J. Am. Soc. Nephrol 27, 1288–1299 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zuk A. & Bonventre JV Acute kidney injury. Annu. Rev. Med 67, 293–307 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Venkatachalam MA, Weinberg JM, Kriz W. & Bidani AK Failed tubule recovery, AKI-CKD transition, and kidney disease progression. J. Am. Soc. Nephrol 26, 1765–1776 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.He L. et al. AKI on CKD: heightened injury, suppressed repair, and the underlying mechanisms. Kidney Int. 92, 1071–1083 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chawla LS, Eggers PW, Star RA & Kimmel PL Acute kidney injury and chronic kidney disease as interconnected syndromes. N. Engl. J. Med 371, 58–66 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ferenbach DA & Bonventre JV Mechanisms of maladaptive repair after AKI leading to accelerated kidney ageing and CKD. Nat. Rev. Nephrol 11, 264–276 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wing MR, Ramezani A, Gill HS, Devaney JM & Raj DS Epigenetics of progression of chronic kidney disease: fact or fantasy? Semin. Nephrol 33, 363–374 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wanner N. & Bechtel-Walz W. Epigenetics of kidney disease. Cell Tissue Res. 369, 75–92 (2017). [DOI] [PubMed] [Google Scholar]
- 13.Bechtel W. et al. Methylation determines fibroblast activation and fibrogenesis in the kidney. Nat. Med 16, 544–550 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Tampe B. et al. Induction of Tet3-dependent epigenetic remodeling by low-dose hydralazine attenuates progression of chronic kidney disease. EBioMedicine 2, 19–36 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Tampe B. et al. Tet3-mediated hydroxymethylation of epigenetically silenced genes contributes to bone morphogenic protein 7-induced reversal of kidney fibrosis. J. Am. Soc. Nephrol 25, 905–912 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Yin S. et al. TGFβ-incurred epigenetic aberrations of miRNA and DNA methyltransferase suppress Klotho and potentiate renal fibrosis. Biochim. Biophys. Acta 1864, 1207–1216 (2017). [DOI] [PubMed] [Google Scholar]
- 17.Xu X. et al. High-fidelity CRISPR/Cas9- based gene-specific hydroxymethylation rescues gene expression and attenuates renal fibrosis. Nat. Commun 9, 3509 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kimura H. Histone modifications for human epigenome analysis. J. Hum. Genet 58, 439–445 (2013). [DOI] [PubMed] [Google Scholar]
- 19.Chen R, Kang R, Fan XG & Tang D. Release and activity of histone in diseases. Cell Death Dis. 5, e1370 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Bhaumik SR, Smith E. & Shilatifard A. Covalent modifications of histones during development and disease pathogenesis. Nat. Struct. Mol. Biol 14, 1008–1016 (2007). [DOI] [PubMed] [Google Scholar]
- 21.Graff J. & Tsai LH Histone acetylation: molecular mnemonics on the chromatin. Nat. Rev. Neurosci 14, 97–111 (2013). [DOI] [PubMed] [Google Scholar]
- 22.Haberland M, Montgomery RL & Olson EN The many roles of histone deacetylases in development and physiology: implications for disease and therapy. Nat. Rev. Genet 10, 32–42 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Greer EL & Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat. Rev. Genet 13, 343–357 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sims RJ III, Nishioka K. & Reinberg D. Histone lysine methylation: a signature for chromatin function. Trends Genet. 19, 629–639 (2003). [DOI] [PubMed] [Google Scholar]
- 25.Kooistra SM & Helin K. Molecular mechanisms and potential functions of histone demethylases. Nat. Rev. Mol. Cell Biol 13, 297–311 (2012). [DOI] [PubMed] [Google Scholar]
- 26.Rossetto D, Avvakumov N. & Cote J. Histone phosphorylation: a chromatin modification involved in diverse nuclear events. Epigenetics 7, 1098–1108 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhang Y, Griffin K, Mondal N. & Parvin JD Phosphorylation of histone H2A inhibits transcription on chromatin templates. J. Biol. Chem 279, 21866–21872 (2004). [DOI] [PubMed] [Google Scholar]
- 28.Wendt KD & Shilatifard A. Packing for the germy: the role of histone H4 Ser1 phosphorylation in chromatin compaction and germ cell development. Genes Dev. 20, 2487–2491 (2006). [DOI] [PubMed] [Google Scholar]
- 29.Marumo T, Hishikawa K, Yoshikawa M. & Fujita T. Epigenetic regulation of BMP7 in the regenerative response to ischemia. J. Am. Soc. Nephrol 19, 1311–1320 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zager RA, Johnson AC & Becker K. Acute unilateral ischemic renal injury induces progressive renal inflammation, lipid accumulation, histone modification, and “end–stage” kidney disease. Am. J. Physiol. Renal Physiol 301, F1334–F1345 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Havasi A. et al. Histone acetyl transferase (HAT) HBO1 and JADE1 in epithelial cell regeneration. Am. J. Pathol 182, 152–162 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Naito M, Bomsztyk K. & Zager RA Renal ischemia-induced cholesterol loading: transcription factor recruitment and chromatin remodeling along the HMG CoA reductase gene. Am. J. Pathol 174, 54–62 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Johnson AC, Ware LB, Himmelfarb J. & Zager RA HMG-CoA reductase activation and urinary pellet cholesterol elevations in acute kidney injury. Clin. J. Am. Soc. Nephrol 6, 2108–2113 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Li HF, Cheng CF, Liao WJ, Lin H. & Yang RB ATF3-mediated epigenetic regulation protects against acute kidney injury. J. Am. Soc. Nephrol 21, 1003–1013 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kim JI, Jung KJ, Jang HS & Park KM Gender-specific role of HDAC11 in kidney ischemia- and reperfusion-induced PAI-1 expression and injury. Am. J. Physiol. Renal Physiol 305, F61–F70 (2013). [DOI] [PubMed] [Google Scholar]
- 36.Hsing CH et al. α2-Adrenoceptor agonist dexmedetomidine protects septic acute kidney injury through increasing BMP-7 and inhibiting HDAC2 and HDAC5. Am. J. Physiol. Renal Physiol 303, F1443–F1453 (2012). [DOI] [PubMed] [Google Scholar]
- 37.Huang J. et al. Histone acetyltransferase PCAF regulates inflammatory molecules in the development of renal injury. Epigenetics 10, 62–72 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Portilla D. et al. Alterations of PPARα and its coactivator PGC-1 in cisplatin-induced acute renal failure. Kidney Int. 62, 1208–1218 (2002). [DOI] [PubMed] [Google Scholar]
- 39.Tran M. et al. PGC–1α promotes recovery after acute kidney injury during systemic inflammation in mice. J. Clin. Invest 121, 4003–4014 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Stallons LJ, Whitaker RM & Schnellmann RG Suppressed mitochondrial biogenesis in folic acid-induced acute kidney injury and early fibrosis. Toxicol. Lett 224, 326–332 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ruiz-Andres O. et al. The inflammatory cytokine TWEAK decreases PGC-1α expression and mitochondrial function in acute kidney injury. Kidney Int. 89, 399–410 (2016). [DOI] [PubMed] [Google Scholar]
- 42.Moreno JA et al. The inflammatory cytokines TWEAK and TNFalpha reduce renal klotho expression through NFκB. J. Am. Soc. Nephrol 22, 1315–1325 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sakao Y. et al. Cisplatin induces Sirt1 in association with histone deacetylation and increased Werner syndrome protein in the kidney. Clin. Exp. Nephrol 15, 363–372 (2011). [DOI] [PubMed] [Google Scholar]
- 44.Hasegawa K. et al. Kidney-specific overexpression of Sirt1 protects against acute kidney injury by retaining peroxisome function. J. Biol. Chem 285, 13045–13056 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.He W. et al. Sirt1 activation protects the mouse renal medulla from oxidative injury. J. Clin. Invest 120, 1056–1068 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dong G. et al. Induction of apoptosis in renal tubular cells by histone deacetylase inhibitors, a family of anticancer agents. J. Pharmacol. Exp. Ther 325, 978–984 (2008). [DOI] [PubMed] [Google Scholar]
- 47.Arany I, Herbert J, Herbert Z. & Safirstein RL Restoration of CREB function ameliorates cisplatin cytotoxicity in renal tubular cells. Am. J. Physiol. Renal Physiol 294, F577–F581 (2008). [DOI] [PubMed] [Google Scholar]
- 48.Dong G, Luo J, Kumar V. & Dong Z. Inhibitors of histone deacetylases suppress cisplatin-induced p53 activation and apoptosis in renal tubular cells. Am. J. Physiol. Renal Physiol 298, F293–F300 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ranganathan P. et al. Histone deacetylase-mediated silencing of AMWAP expression contributes to cisplatin nephrotoxicity. Kidney Int. 89, 317–326 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Liu J. et al. Histone deacetylase inhibitors protect against cisplatin-induced acute kidney injury by activating autophagy in proximal tubular cells. Cell Death Dis. 9, 322 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Yang Y. et al. Renoprotective approaches and strategies in acute kidney injury. Pharmacol. Ther 163, 58–73 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Havasi A. & Dong Z. Autophagy and tubular cell death in the kidney. Semin. Nephrol 36, 174–188 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Pang M. et al. Inhibition of histone deacetylase activity attenuates renal fibroblast activation and interstitial fibrosis in obstructive nephropathy. Am. J. Physiol. Renal Physiol 297, F996–F1005 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Levine MH et al. Class-specific histone/protein deacetylase inhibition protects against renal ischemia reperfusion injury and fibrosis formation. Am. J. Transplant 15, 965–973 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zacharias N. et al. Histone deacetylase inhibitors prevent apoptosis following lethal hemorrhagic shock in rodent kidney cells. Resuscitation 82, 105–109 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Van Beneden K. et al. Valproic acid attenuates proteinuria and kidney injury. J. Am. Soc. Nephrol 22, 1863–1875 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Costalonga EC, Silva FM & Noronha IL Valproic acid prevents renal dysfunction and inflammation in the ischemia–reperfusion injury model. Biomed. Res. Int 2016, 5985903 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Tang J. et al. Class I HDAC activity is required for renal protection and regeneration after acute kidney injury. Am. J. Physiol. Renal Physiol 307, F303–F316 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Cianciolo Cosentino C. et al. Histone deacetylase inhibitor enhances recovery after AKI. J. Am. Soc. Nephrol 24, 943–953 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Novitskaya T. et al. A PTBA small molecule enhances recovery and reduces postinjury fibrosis after aristolochic acid–induced kidney injury. Am. J. Physiol. Renal Physiol 306, F496–F504 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Shi Y. et al. Inhibition of HDAC6 protects against rhabdomyolysis-induced acute kidney injury. Am. J. Physiol. Renal Physiol 312, F502–F515 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Tang J. et al. Blockade of histone deacetylase 6 protects against cisplatin-induced acute kidney injury. Clin. Sci. (Lond.) 132, 339–359 (2018). [DOI] [PubMed] [Google Scholar]
- 63.Do Amaral CL et al. Resveratrol attenuates cisplatin-induced nephrotoxicity in rats. Arch. Toxicol 82, 363–370 (2008). [DOI] [PubMed] [Google Scholar]
- 64.Xu S. et al. SIRT1/3 activation by resveratrol attenuates acute kidney injury in a septic rat model. Oxid. Med. Cell Longev 2016, 7296092 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Fan H. et al. The histone deacetylase, SIRT1, contributes to the resistance of young mice to ischemia/reperfusion-induced acute kidney injury. Kidney Int. 83, 404–413 (2013). [DOI] [PubMed] [Google Scholar]
- 66.Kuhad A, Pilkhwal S, Sharma S, Tirkey N. & Chopra K. Effect of curcumin on inflammation and oxidative stress in cisplatin-induced experimental nephrotoxicity. J. Agr. Food Chem 55, 10150–10155 (2007). [DOI] [PubMed] [Google Scholar]
- 67.Bayrak O. et al. Curcumin protects against ischemia/reperfusion injury in rat kidneys. World J. Urol 26, 285–291 (2008). [DOI] [PubMed] [Google Scholar]
- 68.Memis D. et al. Curcumin attenuates the organ dysfunction caused by endotoxemia in the rat. Nutrition 24, 1133–1138 (2008). [DOI] [PubMed] [Google Scholar]
- 69.Glozak MA, Sengupta N, Zhang X. & Seto E. Acetylation and deacetylation of non-histone proteins. Gene 363, 15–23 (2005). [DOI] [PubMed] [Google Scholar]
- 70.Naito M, Bomsztyk K. & Zager RA Endotoxin mediates recruitment of RNA polymerase II to target genes in acute renal failure. J. Am. Soc. Nephrol 19, 1321–1330 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Munshi R. et al. MCP-1 gene activation marks acute kidney injury. J. Am. Soc. Nephrol 22, 165–175 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Zhou X. et al. Enhancer of zeste homolog 2 inhibition attenuates renal fibrosis by maintaining Smad7 and phosphatase and tensin homolog expression. J. Am. Soc. Nephrol 27, 2092–2108 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Zhou X. et al. Targeting histone methyltransferase enhancer of zeste homolog-2 inhibits renal epithelial-mesenchymal transition and attenuates renal fibrosis. FASEB J. 32, fj201800237R (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mimura I. et al. Genome-wide analysis revealed that DZNep reduces tubulointerstitial fibrosis via downregulation of pro-fibrotic genes. Sci. Rep 8, 3779 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Tikoo K, Lau SS & Monks TJ Histone H3 phosphorylation is coupled to poly-(ADP-ribosylation) during reactive oxygen species-induced cell death in renal proximal tubular epithelial cells. Mol. Pharmacol 60, 394–402 (2001). [DOI] [PubMed] [Google Scholar]
- 76.Pabla N, Huang S, Mi QS, Daniel R. & Dong Z. ATR-Chk2 signaling in p53 activation and DNA damage response during cisplatin-induced apoptosis. J. Biol. Chem 283, 6572–6583 (2008). [DOI] [PubMed] [Google Scholar]
- 77.Ma Z, Wei Q, Dong G, Huo Y. & Dong Z. DNA damage response in renal ischemia-reperfusion and ATP-depletion injury of renal tubular cells. Biochim. Biophys. Acta 1842, 1088–1096 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Scholpa NE, Zhang X, Kolli RT & Cummings BS Epigenetic changes in p21 expression in renal cells after exposure to bromate. Toxicol. Sci 141, 432–440 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zhu S, Pabla N, Tang C, He L. & Dong Z. DNA damage response in cisplatin-induced nephrotoxicity. Arch. Toxicol 89, 2197–2205 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Sabari BR et al. Intracellular crotonyl-CoA stimulates transcription through p300-catalyzed histone crotonylation. Mol. Cell 58, 203–215 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ruiz-Andres O. et al. Histone lysine crotonylation during acute kidney injury in mice. Dis. Model. Mech 9, 633–645 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Mar D. et al. Heterogeneity of epigenetic changes at ischemia/reperfusion- and endotoxin-induced acute kidney injury genes. Kidney Int. 88, 734–744 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Schubeler D. Function and information content of DNA methylation. Nature 517, 321–326 (2015). [DOI] [PubMed] [Google Scholar]
- 84.Jeltsch A. Beyond Watson and Crick: DNA methylation and molecular enzymology of DNA methyltransferases. Chembiochem 3, 274–293 (2002). [DOI] [PubMed] [Google Scholar]
- 85.Ramsahoye BH et al. Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc. Natl Acad. Sci. USA 97, 5237–5242 (2000). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Lister R. et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462, 315–322 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Shirane K. et al. Mouse oocyte methylomes at base resolution reveal genome-wide accumulation of non-CpG methylation and role of DNA methyltransferases. PLOS Genet. 9, e1003439 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Guo JU et al. Distribution, recognition and regulation of non-CpG methylation in the adult mammalian brain. Nat. Neurosci 17, 215–222 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Xie W. et al. Base-resolution analyses of sequence and parent-of-origin dependent DNA methylation in the mouse genome. Cell 148, 816–831 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Messerschmidt DM, Knowles BB & Solter D. DNA methylation dynamics during epigenetic reprogramming in the germline and preimplantation embryos. Genes Dev. 28, 812–828 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Portela A. & Esteller M. Epigenetic modifications and human disease. Nat. Biotechnol 28, 1057–1068 (2010). [DOI] [PubMed] [Google Scholar]
- 92.Li E. & Zhang Y. DNA methylation in mammals. Cold Spring Harb. Perspect. Biol 6, a019133 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Rodriguez J. et al. Chromosomal instability correlates with genome-wide DNA demethylation in human primary colorectal cancers. Cancer Res. 66, 8462–8468 (2006). [DOI] [PubMed] [Google Scholar]
- 94.Song F. et al. Association of tissue-specific differentially methylated regions (TDMs) with differential gene expression. Proc. Natl Acad. Sci. USA 102, 3336–3341 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Weber M. et al. Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells. Nat. Genet 37, 853–862 (2005). [DOI] [PubMed] [Google Scholar]
- 96.Okano M, Xie S. & Li E. Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nat. Genet 19, 219–220 (1998). [DOI] [PubMed] [Google Scholar]
- 97.Gowher H, Liebert K, Hermann A, Xu G. & Jeltsch A. Mechanism of stimulation of catalytic activity of Dnmt3A and Dnmt3B DNA-(cytosine-C5)-methyltransferases by Dnmt3L. J. Biol. Chem 280, 13341–13348 (2005). [DOI] [PubMed] [Google Scholar]
- 98.Goll MG et al. Methylation of tRNAAsp by the DNA methyltransferase homolog Dnmt2. Science 311, 395–398 (2006). [DOI] [PubMed] [Google Scholar]
- 99.Schaefer M. et al. RNA methylation by Dnmt2 protects transfer RNAs against stress-induced cleavage. Genes Dev. 24, 1590–1595 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Li E, Bestor TH & Jaenisch R. Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell 69, 915–926 (1992). [DOI] [PubMed] [Google Scholar]
- 101.Okano M, Bell DW, Haber DA & Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 99, 247–257 (1999). [DOI] [PubMed] [Google Scholar]
- 102.Bogdanovic O. & Veenstra GJ DNA methylation and methyl-CpG binding proteins: developmental requirements and function. Chromosoma 118, 549–565 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Moore LD, Le T. & Fan G. DNA methylation and its basic function. Neuropsychopharmacology 38, 23–38 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Nan X. et al. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 393, 386–389 (1998). [DOI] [PubMed] [Google Scholar]
- 105.Ng HH et al. MBD2 is a transcriptional repressor belonging to the MeCP1 histone deacetylase complex. Nat. Genet 23, 58–61 (1999). [DOI] [PubMed] [Google Scholar]
- 106.Filion GJ et al. A family of human zinc finger proteins that bind methylated DNA and repress transcription. Mol. Cell. Biol 26, 169–181 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Wu SC & Zhang Y. Active DNA demethylation: many roads lead to Rome. Nat. Rev. Mol. Cell Biol 11, 607–620 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Ramiro AR & Barreto VM Activation-induced cytidine deaminase and active cytidine demethylation. Trends Biochem. Sci 40, 172–181 (2015). [DOI] [PubMed] [Google Scholar]
- 109.Kohli RM & Zhang Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature 502, 472–479 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Pastor WA, Aravind L. & Rao A. TETonic shift: biological roles of TET proteins in DNA demethylation and transcription. Nat. Rev. Mol. Cell Biol 14, 341–356 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Guo JU, Su Y, Zhong C, Ming GL & Song H. Hydroxylation of 5-methylcytosine by TET1 promotes active DNA demethylation in the adult brain. Cell 145, 423–434 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Dawlaty MM et al. Tet1 is dispensable for maintaining pluripotency and its loss is compatible with embryonic and postnatal development. Cell Stem Cell 9, 166–175 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Ko M. et al. Ten-eleven-translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice. Proc. Natl Acad. Sci. USA 108, 14566–14571 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Dawlaty MM et al. Combined deficiency of Tet1 and Tet2 causes epigenetic abnormalities but is compatible with postnatal development. Dev. Cell 24, 310–323 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Gu TP et al. The role of Tet3 DNA dioxygenase in epigenetic reprogramming by oocytes. Nature 477, 606–610 (2011). [DOI] [PubMed] [Google Scholar]
- 116.Smyth LJ, McKay GJ, Maxwell AP & McKnight AJ DNA hypermethylation and DNA hypomethylation is present at different loci in chronic kidney disease. Epigenetics 9, 366–376 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Wing MR et al. DNA methylation profile associated with rapid decline in kidney function: findings from the CRIC study. Nephrol. Dial. Transplant 29, 864–872 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Ko YA et al. Cytosine methylation changes in enhancer regions of core pro-fibrotic genes characterize kidney fibrosis development. Genome Biol. 14, R108 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Marumo T. et al. Diabetes induces aberrant DNA methylation in the proximal tubules of the kidney. J. Am. Soc. Nephrol 26, 2388–2397 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Brennan EP et al. DNA methylation profiling in cell models of diabetic nephropathy. Epigenetics 5, 396–401 (2010). [DOI] [PubMed] [Google Scholar]
- 121.Pratt JR et al. Ischemic epigenetics and the transplanted kidney. Transplant. Proc 38, 3344–3346 (2006). [DOI] [PubMed] [Google Scholar]
- 122.Parker MD, Chambers PA, Lodge JP & Pratt JR Ischemia-reperfusion injury and its influence on the epigenetic modification of the donor kidney genome. Transplantation 86, 1818–1823 (2008). [DOI] [PubMed] [Google Scholar]
- 123.Endo K, Kito N, Fukushima Y, Weng H. & Iwai N. A novel biomarker for acute kidney injury using TaqMan-based unmethylated DNA-specific polymerase chain reaction. Biomed. Res 35, 207–213 (2014). [DOI] [PubMed] [Google Scholar]
- 124.Kang SW et al. Renal kallikrein excretion and epigenetics in human acute kidney injury: expression, mechanisms and consequences. BMC Nephrol. 12, 27 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Mehta TK et al. Quantitative detection of promoter hypermethylation as a biomarker of acute kidney injury during transplantation. Transplant. Proc 38, 3420–3426 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Huang N, Tan L, Xue Z, Cang J. & Wang H. Reduction of DNA hydroxymethylation in the mouse kidney insulted by ischemia reperfusion. Biochem. Biophys. Res. Commun 422, 697–702 (2012). [DOI] [PubMed] [Google Scholar]
- 127.Wang H, Huang N, Liu Y, Cang J. & Xue Z. Genomic distribution of 5-Hydroxymethylcytosine in mouse kidney and its relationship with gene expression. Ren. Fail 38, 982–988 (2016). [DOI] [PubMed] [Google Scholar]
- 128.Zhao Y. et al. Genome-wide DNA methylation analysis in renal ischemia reperfusion injury. Gene 610, 32–43 (2017). [DOI] [PubMed] [Google Scholar]
- 129.Guo C. et al. DNA methylation protects against cisplatin-induced kidney injury by regulating specific genes, including interferon regulatory factor 8. Kidney Int. 92, 1194–1205 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Branco MR, Ficz G. & Reik W. Uncovering the role of 5-hydroxymethylcytosine in the epigenome. Nat. Rev. Genet 13, 7–13 (2011). [DOI] [PubMed] [Google Scholar]
- 131.Globisch D. et al. Tissue distribution of 5-hydroxymethylcytosine and search for active demethylation intermediates. PLOS ONE 5, e15367 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Chu AY et al. Epigenome-wide association studies identify DNA methylation associated with kidney function. Nat. Commun 8, 1286 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Tampe B. et al. Low-dose hydralazine prevents fibrosis in a murine model of acute kidney injury-to-chronic kidney disease progression. Kidney Int. 91, 157–176 (2017). [DOI] [PubMed] [Google Scholar]
- 134.Hu MC & Moe OW Klotho as a potential biomarker and therapy for acute kidney injury. Nat. Rev. Nephrol 8, 423–429 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Mencke R, Olauson H. & Hillebrands JL Effects of Klotho on fibrosis and cancer: a renal focus on mechanisms and therapeutic strategies. Adv. Drug Deliv. Rev 121, 85–100 (2017). [DOI] [PubMed] [Google Scholar]
- 136.Chen J. et al. Elevated Klotho promoter methylation is associated with severity of chronic kidney disease. PLOS ONE 8, e79856 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Zhang Q, Yin S, Liu L, Liu Z. & Cao W. Rhein reversal of DNA hypermethylation-associated Klotho suppression ameliorates renal fibrosis in mice. Sci. Rep 6, 34597 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Tirkey N, Kaur G, Vij G. & Chopra K. Curcumin, a diferuloylmethane, attenuates cyclosporine-induced renal dysfunction and oxidative stress in rat kidneys. BMC Pharmacol. 5, 15 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Zhang Q. et al. Rhein reverses Klotho repression via promoter demethylation and protects against kidney and bone injuries in mice with chronic kidney disease. Kidney Int. 91, 144–156 (2017). [DOI] [PubMed] [Google Scholar]
- 140.Bartel DP MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 116, 281–297 (2004). [DOI] [PubMed] [Google Scholar]
- 141.Chua JH, Armugam A. & Jeyaseelan K.MicroRNAs: biogenesis, function and applications. Curr. Opin. Mo!. Ther 11, 189–199 (2009). [PubMed] [Google Scholar]
- 142.Wang KC & Chang HY Molecular mechanisms of long noncoding RNAs. Mol. Cell43, 904–914 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Karlsson O. & Baccarelli AA Environmental health and long non-coding RNAs. Curr. Environ. Health Rep 3, 178–187 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Neguembor MV, Jothi M. & Gabellini D. Long noncoding RNAs, emerging players in muscle differentiation and disease. Skelet. Muscle 4, 8 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Quek XC et al. lncRNAdb v2.0: expanding the reference database for functional long noncoding RNAs. Nucleic Acids Res. 43, D168–D173 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Alvarez-Garcia I. & Miska EA MicroRNA functions in animal development and human disease. Development 132, 4653–4662 (2005). [DOI] [PubMed] [Google Scholar]
- 147.Flynt AS & Lai EC Biological principles of microRNA-mediated regulation: shared themes amid diversity. Nat. Rev. Genet 9, 831–842 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Bushati N. & Cohen SM microRNA functions. Annu. Rev. Cell Dev. Biol 23, 175–205 (2007). [DOI] [PubMed] [Google Scholar]
- 149.Chang TC & Mendell JT microRNAs in vertebrate physiology and human disease. Annu. Rev. Genomics Hum. Genet 8, 215–239 (2007). [DOI] [PubMed] [Google Scholar]
- 150.Mendell JT & Olson EN MicroRNAs in stress signaling and human disease. Cell 148, 1172–1187 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Erson AE & Petty EM MicroRNAs in development and disease. Clin. Genet 74, 296–306 (2008). [DOI] [PubMed] [Google Scholar]
- 152.Tufekci KU, Oner MG, Meuwissen RL & Genc S. The role of microRNAs in human diseases. Methods Mol. Biol 1107, 33–50 (2014). [DOI] [PubMed] [Google Scholar]
- 153.Ullah S, John P. & Bhatti A. MicroRNAs with a role in gene regulation and in human diseases. Mol. Biol. Rep 41, 225–232 (2014). [DOI] [PubMed] [Google Scholar]
- 154.Karolina DS, Wintour EM, Bertram J. & Jeyaseelan K. Riboregulators in kidney development and function. Biochimie 92, 217–225 (2010). [DOI] [PubMed] [Google Scholar]
- 155.Saal S. & Harvey SJ MicroRNAs and the kidney: coming of age. Curr. Opin. Nephrol. Hypertens 18, 317–323 (2009). [DOI] [PubMed] [Google Scholar]
- 156.Wei Q, Mi QS & Dong Z. The regulation and function of microRNAs in kidney diseases. IUBMB Life 65, 602–614 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Bhatt K, Mi QS & Dong Z. microRNAs in kidneys: biogenesis, regulation, and pathophysiological roles. Am. J. Physiol. Renal Physiol 300, F602–F610 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Ho J. & Kreidberg JA The long and short of microRNAs in the kidney. J. Am. Soc. Nephrol 23, 400–404 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Chandrasekaran K. et al. Role of microRNAs in kidney homeostasis and disease. Kidney Int. 81, 617–627 (2012). [DOI] [PubMed] [Google Scholar]
- 160.Ho J. & Kreidberg JA MicroRNAs in renal development. Pediatr. Nephrol 28, 219–225 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Khalid U, Bowen T, Fraser DJ & Jenkins RH Acute kidney injury: a paradigm for miRNA regulation of the cell cycle. Biochem. Soc. Trans 42, 1219–1223 (2014). [DOI] [PubMed] [Google Scholar]
- 162.Aguado-Fraile E. et al. MicroRNAs in the kidney: novel biomarkers of acute kidney injury. Nefrologia 33, 826–834 (2013). [DOI] [PubMed] [Google Scholar]
- 163.Liu Z, Wang S, Mi QS & Dong Z. MicroRNAs in pathogenesis of acute kidney injury. Nephron 134, 149–153 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Ma L. & Qu L. The function of microRNAs in renal development and pathophysiology. J. Genet. Genomics 40, 143–152 (2013). [DOI] [PubMed] [Google Scholar]
- 165.Trionfini P, Benigni A. & Remuzzi G. MicroRNAs in kidney physiology and disease. Nat. Rev. Nephrol 11, 23–33 (2015). [DOI] [PubMed] [Google Scholar]
- 166.Mukhadi S, Hull R, Mbita Z. & Dlamini Z. The role of MicroRNAs in kidney disease. Noncoding RNA 1, 192–221 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Bhatt K, Kato M. & Natarajan R. Mini-review: emerging roles of microRNAs in the pathophysiology of renal diseases. Am. J. Physiol. Renal Physiol 310, F109–F118 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Wei Q. et al. Targeted deletion of Dicer from proximal tubules protects against renal ischemia-reperfusion injury. J. Am. Soc. Nephrol 21, 756–761 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Godwin JG et al. Identification of a microRNA signature of renal ischemia reperfusion injury.Proc. Natl Acad. Sci. USA 107, 14339–14344 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Shapiro MD et al. MicroRNA expression data reveals a signature of kidney damage following ischemia reperfusion injury. PLOS ONE 6, e23011 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Pellegrini KL et al. Application of small RNA sequencing to identify microRNAs in acute kidney injury and fibrosis. Toxicol. Appl. Pharmacol 312, 42–52 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Lorenzen JM et al. MicroRNA-24 antagonism prevents renal ischemia reperfusion injury. J. Am. Soc. Nephrol 25, 2717–2729 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Ranganathan P. et al. MicroRNA-150 deletion in mice protects kidney from myocardial infarction-induced acute kidney injury. Am. J. Physiol. Renal Physiol 309, F551–F558 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Lan YF et al. MicroRNA-494 reduces ATF3 expression and promotes AKI. J. Am. Soc. Nephrol 23, 2012–2023 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Bhatt K. et al. MicroRNA-687 induced by hypoxia-inducible factor-1 targets phosphatase and tensin homolog in renal ischemia-reperfusion injury. J. Am. Soc. Nephrol 26, 1588–1596 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Hao J. et al. Induction of microRNA-17–5p by p53 protects against renal ischemia-reperfusion injury by targeting death receptor 6. Kidney Int. 91, 106–118 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Liang S, Wang W. & Gou X. MicroRNA 26a modulates regulatory T cells expansion and attenuates renal ischemia-reperfusion injury. Mol. Immunol 65, 321–327 (2015). [DOI] [PubMed] [Google Scholar]
- 178.Bijkerk R. et al. Hematopoietic microRNA-126 protects against renal ischemia/reperfusion injury by promoting vascular integrity. J. Am. Soc. Nephrol 25, 1710–1722 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Aguado-Fraile E. et al. miR-127 protects proximal tubule cells against ischemia/reperfusion: identification of kinesin family member 3B as miR–127 target. PLOS ONE 7, e44305 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Dai Y. et al. miR-146a is essential for lipopolysaccharide (LPS)-induced cross-tolerance against kidney ischemia/reperfusion injury in mice. Sci. Rep 6, 27091 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Amrouche L. et al. MicroRNA-146a in human and experimental ischemic AKI: CXCL8-dependent mechanism of action. J. Am. Soc. Nephrol 28, 479–493 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Muratsu-Ikeda S. et al. Downregulation of miR-205 modulates cell susceptibility to oxidative and endoplasmic reticulum stresses in renal tubular cells. PLOS ONE 7, e41462 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Wei Q. et al. MicroRNA-489 induction by hypoxia-inducible factor-1 protects against ischemic kidney injury. J. Am. Soc. Nephrol 27, 2784–2796 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Xu X. et al. Delayed ischemic preconditioning contributes to renal protection by upregulation of miR-21. Kidney Int. 82, 1167–1175 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Jia P. et al. miR-21 contributes to xenon-conferred amelioration of renal ischemia-reperfusion injury in mice. Anesthesiology 119, 621–630 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.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 40, 215–223 (2014). [DOI] [PubMed] [Google Scholar]
- 187.Xu X. et al. Renal protection mediated by hypoxia inducible factor-1α depends on proangiogenesis function of miR-21 by targeting thrombospondin 1. Transplantation 101, 1811–1819 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Liu X. et al. MiR-21 inhibits autophagy by targeting Rab11a in renal ischemia/reperfusion. Exp. Cell Res 338, 64–69 (2015). [DOI] [PubMed] [Google Scholar]
- 189.El Gazzar M, Church A, Liu T. & McCall CE MicroRNA-146a regulates both transcription silencing and translation disruption of TNF-α during TLR4- induced gene reprogramming. J. Leukoc. Biol 90, 509–519 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Saba R, Sorensen DL & Booth SA MicroRNA-146a: a dominant, negative regulator of the innate immune response. Front. Immunol 5, 578 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Bhatt K. et al. MicroRNA-34a is induced via p53 during cisplatin nephrotoxicity and contributes to cell survival. Mol. Med 16, 409–416 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Lee CG et al. Discovery of an integrative network of microRNAs and transcriptomics changes for acute kidney injury. Kidney Int. 86, 943–953 (2014). [DOI] [PubMed] [Google Scholar]
- 193.Joo MS, Lee CG, Koo JH & Kim SG miR-125b transcriptionally increased by Nrf2 inhibits AhR repressor, which protects kidney from cisplatin-induced injury. Cell Death Dis. 4, e899 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Zhu Y. et al. MicroRNA-146b, a sensitive indicator of mesenchymal stem cell repair of acute renal injury. Stem Cells Transl Med. 5, 1406–1415 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Pellegrini KL et al. MicroRNA-155 deficient mice experience heightened kidney toxicity when dosed with cisplatin. Toxicol. Sci 141, 484–492 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Zhu HY et al. Role of microRNA-181a in the apoptosis of tubular epithelial cell induced by cisplatin. Chin. Med. J 125, 523–526 (2012). [PubMed] [Google Scholar]
- 197.Qin W, Xie W, Yang X, Xia N. & Yang K. Inhibiting microRNA-449 attenuates cisplatin-induced injury in NRK-52E cells possibly via regulating the SIRT1/P53/BAX pathway. Med. Sci. Monit 22, 818–823 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Hao J. et al. MicroRNA-375 is induced in cisplatin nephrotoxicity to repress hepatocyte nuclear factor 1-β. J. Biol. Chem 292, 4571–4582 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Guo Y. et al. MicroRNA-709 mediates acute tubular injury through effects on mitochondrial function. J. Am. Soc. Nephrol 29, 449–461 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Jenkins RH et al. miR-192 induces G2/M growth arrest in aristolochic acid nephropathy. Am. J. Pathol 184, 996–1009 (2014). [DOI] [PubMed] [Google Scholar]
- 201.Weber JA et al. The microRNA spectrum in 12 body fluids. Clin. Chem 56, 1733–1741 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Lorenzen JM et al. Circulating miR-210 predicts survival in critically ill patients with acute kidney injury. Clin. J. Am. Soc. Nephrol 6, 1540–1546 (2011). [DOI] [PubMed] [Google Scholar]
- 203.Wang N. et al. Urinary microRNA-10a and microRNA-30d serve as novel, sensitive and specific biomarkers for kidney injury. PLOS ONE 7, e51140 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Saikumar J. et al. Expression, circulation, and excretion profile of microRNA-21, −155, and −18a following acute kidney injury. Toxicol. Sci 129, 256–267 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Du J. et al. MicroRNA-21 and risk of severe acute kidney injury and poor outcomes after adult cardiac surgery. PLOS ONE 8, e63390 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Ramachandran K. et al. Human miRNome profiling identifies microRNAs differentially present in the urine after kidney injury. Clin. Chem 59, 1742–1752 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Kanki M. et al. Identification of urinary miRNA biomarkers for detecting cisplatin-induced proximal tubular injury in rats. Toxicology 324, 158–168 (2014). [DOI] [PubMed] [Google Scholar]
- 208.Wang JF et al. Screening plasma miRNAs as biomarkers for renal ischemia-reperfusion injury in rats. Med. Sci. Monit 20, 283–289 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Pavkovic M, Riefke B. & Ellinger-Ziegelbauer H. Urinary microRNA profiling for identification of biomarkers after cisplatin-induced kidney injury. Toxicology 324, 147–157 (2014). [DOI] [PubMed] [Google Scholar]
- 210.Bellinger MA et al. Concordant changes of plasma and kidney microRNA in the early stages of acute kidney injury: time course in a mouse model of bilateral renal ischemia-reperfusion. PLOS ONE 9, e93297 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Aguado-Fraile E. et al. A pilot study identifying a set of microRNAs as precise diagnostic biomarkers of acute kidney injury. PLOS ONE 10, e0127175 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Kito N, Endo K, Ikesue M, Weng H. & Iwai N. miRNA profiles of tubular cells: diagnosis of kidney injury. Biomed. Res. Int 2015, 465479 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Tsai PC et al. Serum microRNA-21 and microRNA-221 as potential biomarkers for cerebrovascular disease. J. Vasc. Res 50, 346–354 (2013). [DOI] [PubMed] [Google Scholar]
- 214.Szeto CC et al. Micro–RNA expression in the urinary sediment of patients with chronic kidney diseases. Dis. Markers 33, 137–144 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Yuan XP et al. MicroRNA-423–5p facilitates hypoxia/reoxygenation-induced apoptosis in renal proximal tubular epithelial cells by targeting GSTM1 via endoplasmic reticulum stress. Oncotarget 8, 82064–82077 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Dreval K. et al. miR-1247 blocks SOX9-mediated regeneration in alcohol- and fibrosis-associated acute kidney injury in mice. Toxicology 384, 40–49 (2017). [DOI] [PubMed] [Google Scholar]
- 217.Lin J. et al. The long noncoding RNA landscape in hypoxic and inflammatory renal epithelial injury. Am. J. Physiol. Renal Physiol 309, F901–F913 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Mimura I. et al. Novel lnc RNA regulated by HIF-1 inhibits apoptotic cell death in the renal tubular epithelial cells under hypoxia. Physiol. Rep 5, e13203 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Yu TM et al. RANTES mediates kidney ischemia reperfusion injury through a possible role of HIF-1α and LncRNA PRINS. Sci. Rep 6, 18424 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Lelli A. et al. Induction of long noncoding RNA MALAT1 in hypoxic mice. Hypoxia (Auckl.) 3, 45–52 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Kolling M. et al. Hypoxia-induced long non-coding RNA Malatl is dispensable for renal ischemia/reperfusion-injury. Sci. Rep 8, 3438 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Geng X. et al. The effect of long noncoding RNA GAS5 on apoptosis in renal ischemia/reperfusion injury. Nephrology (Carlton) 10.1111/nep.13476 (2018). [DOI] [Google Scholar]
- 223.Ding Y. et al. Mechanism of long non-coding RNA MALAT1 in lipopolysaccharide-induced acute kidney injury is mediated by the miR-146a/NF-κB signaling pathway. Int. J. Mo!. Med 41, 446–454 (2018). [DOI] [PubMed] [Google Scholar]
- 224.Huang W. et al. Long non-coding RNA PVT1 promote LPS-induced septic acute kidney injury by regulating TNFalpha and JNK/NF-kappaB pathways in HK-2 cells. Int. Immunopharmacol 47, 134–140 (2017). [DOI] [PubMed] [Google Scholar]
- 225.Chen Y. et al. Long non-coding RNA NEAT1 plays an important role in sepsis-induced acute kidney injury by targeting miR-204 and modulating the NF-κB pathway. Int. Immunopharmacol 59, 252–260 (2018). [DOI] [PubMed] [Google Scholar]
- 226.Yang R. et al. Inhibition of maternally expressed gene 3 attenuated lipopolysaccharide-induced apoptosis through sponging miR-21 in renal tubular epithelial cells. J. Cell. Biochem 119, 7800–7806 (2018). [DOI] [PubMed] [Google Scholar]
- 227.Shen J, Zhang J, Jiang X, Wang H. & Pan G. LncRNA HOX transcript antisense RNA accelerated kidney injury induced by urine-derived sepsis through the miR-22/high mobility group Box 1 pathway. Life Sci. 210, 185–191 (2018). [DOI] [PubMed] [Google Scholar]
- 228.Lorenzen JM et al. Circulating long noncoding RNATapSaki is a predictor of mortality in critically ill patients with acute kidney injury. Clin. Chem 61, 191–201 (2015). [DOI] [PubMed] [Google Scholar]
- 229.Arvaniti E. et al. Whole-transcriptome analysis of UUO mouse model of renal fibrosis reveals new molecular players in kidney diseases. Sci. Rep 6, 26235 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Zhou Q. et al. Identification of novel long noncoding RNAs associated with TGF-β/Smad3-mediated renal inflammation and fibrosis by RNA sequencing. Am. J. Pathol 184, 409–417 (2014). [DOI] [PubMed] [Google Scholar]
- 231.Sun J, Zhang S, Shi B, Zheng D. & Shi J. Transcriptome identified lncRNAs associated with renal fibrosis in UUO rat model. Front. Physiol 8, 658 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Feng M. et al. TGF-β mediates renal fibrosis via the Smad3-Erbb4-IR long noncoding RNA axis. Mol. Ther 26, 148–161 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Fontecha-Barriuso M. et al. Targeting epigenetic DNA and histone modifications to treat kidney disease. Nephrol. Dial. Transplant 33, 1875–1886 (2018). [DOI] [PubMed] [Google Scholar]
- 234.Mellis D. & Caporali A. MicroRNA-based therapeutics in cardiovascular disease: screening and delivery to the target. Biochem. Soc. Trans 46, 11–21 (2018). [DOI] [PubMed] [Google Scholar]




