Skip to main content
American Journal of Physiology - Renal Physiology logoLink to American Journal of Physiology - Renal Physiology
. 2015 Nov 4;310(2):F109–F118. doi: 10.1152/ajprenal.00387.2015

Mini-review: emerging roles of microRNAs in the pathophysiology of renal diseases

Kirti Bhatt 1, Mitsuo Kato 1, Rama Natarajan 1,
PMCID: PMC4719041  PMID: 26538441

Abstract

MicroRNAs (miRNA) are endogenously produced short noncoding regulatory RNAs that can repress gene expression by posttranscriptional mechanisms. They can therefore influence both normal and pathological conditions in diverse biological systems. Several miRNAs have been detected in kidneys, where they have been found to be crucial for renal development and normal physiological functions as well as significant contributors to the pathogenesis of renal disorders such as diabetic nephropathy, acute kidney injury, lupus nephritis, polycystic kidney disease, and others, due to their effects on key genes involved in these disease processes. miRNAs have also emerged as novel biomarkers in these renal disorders. Due to increasing evidence of their actions in various kidney segments, in this mini-review we discuss the functional significance of altered miRNA expression during the development of renal pathologies and highlight emerging miRNA-based therapeutics and diagnostic strategies for early detection and treatment of kidney diseases.

Keywords: acute kidney injury, diabetic nephropathy, kidney, microRNA, polycystic kidney disease


micrornas (mirnas/mirs) are a unique class of short (∼22 nucleotides) single-stranded endogenous noncoding RNAs (7, 18). miRNAs contain nucleotide sequences complementary to binding sites on the 3′-untranslated regions (3′-UTRs) of protein-coding messenger RNA (mRNA) transcripts. Through direct binding to the mRNA 3′-UTRs, miRNAs can induce repression of target gene and protein expression (56). Individual miRNAs can often engage numerous target mRNAs, thereby influencing several components of a single or multiple cellular pathways (3). By regulating and modulating gene expression via posttranscriptional mechanisms, miRNAs are now recognized as critical players in a variety of developmental, physiological, and pathological conditions.

miRNA biogenesis, processing, and actions occur by discrete mechanisms (44, 56, 66). miRNA genes are located within various genomic contexts, including introns of noncoding or coding transcripts as well as exonic regions. miRNAs are transcribed by RNA polymerase II as long precursor RNAs, called primary miRNAs (pri-miRNAs), which are sequentially processed by key nucleases in the nucleus and cytoplasm to generate mature miRNAs (44). The initial pri-miRNA transcript contains a local stem-loop structure harboring the mature miRNA sequence. Several extensive maturation steps are required to further process the pri-miRNA transcript, leading to the formation of mature miRNA. Initially, within the nucleus, the microprocessor complex, which includes RNase III Drosha and DiGeorge syndrome chromosomal region 8 (DGCR8) crops the stem-loop in the pri-miRNA, which releases a small hairpin-shaped RNA of ∼65 nucleotides in length, called pre-miRNA. This pre-miRNA is then exported into the cytoplasm by a multiprotein transport complex, which includes exportin 5 and GTP-binding nuclear protein RAN·GTP. In the cytoplasm, pre-miRNA is cleaved by the RNase III-type endonuclease Dicer, which liberates a small RNA duplex. The resultant RNA duplex is subsequently loaded onto argonaute proteins (AGO1-4) in the RNA-induced silencing complex (RISC) machinery, forming a site where the “passenger” strand is degraded while the mature microRNA (miRNA) remains in complex with the AGO proteins. Besides this canonical biogenesis pathway, various alternative mechanisms can also generate miRNAs (44). These biogenesis pathways have been described in depth in several recent review articles (44, 81, 161). Once processed and loaded into the silencing complex, the mature miRNAs pair with target mRNAs, leading to direct translational repression, mRNA destabilization, or a combination of the two.

More than 2,000 miRNA genes have been identified in humans, which are thought to regulate a high percentage of protein-coding transcripts (3). Small variations in the levels of miRNAs can have major cellular effects, and alterations in miRNA expression are observed during progression of various human diseases (98). Notably, functional studies have established miRNAs as critical modulators of several disease conditions (98). As a result, therapeutic targeting of miRNA function through systemic or local delivery of miRNA inhibitors or mimics is being extensively pursued (80). The recent successful clinical trial (53) utilizing a miRNA therapeutic for suppression of hepatitis C virus replication have raised the possibility for developing miRNA-based therapeutics for other diseases as well. Moreover, miRNAs have emerged as prognostic and predictive biomarkers for human diseases owing to their high stability and presence in blood, urine, and other body fluids (29). Along with cancer, cardiovascular, and neurological diseases (98), recent work has revealed the key roles played by miRNAs in the development and progression of kidney diseases (64, 141). These studies have raised the possibility of development of novel diagnostic and therapeutic strategies that can be clinically translated for the prevention and treatment of various renal diseases.

Diabetic Nephropathy

Diabetic nephropathy (DN) is one of the most common and debilitating complications of diabetes (40, 57) that can quite often lead to proteinuria and end-stage renal disease (ESRD) (57). Patients with DN also have a higher risk of macrovascular complications (40, 57, 126). Glomerular mesangial hypertrophy, mesangial expansion, and accumulation of extracellular matrix (ECM) proteins in the glomerular and tubular compartments, as well as podocyte effacement and apoptosis, are major features of DN (23, 60, 64, 100, 107, 120). Although improved glycemic control reduces the rates of microvascular complications including DN (27, 28, 105, 111), the global incidence of DN is still escalating, suggesting that new mediators and mechanisms need to be considered for more effective treatment.

High-glucose (HG) conditions seen during diabetes can adversely affect several renal cells involved in DN and also increase the formation of advanced glycation end products (AGEs) and production of growth factors such as transforming growth factor-β1 (TGF-β1), angiotensin II, and platelet-derived growth factor in renal cells, which can cooperate to contribute to DN pathogenesis (1, 64, 123, 129, 165). In particular, TGF-β1 is a major player in DN pathogenesis, due to its ability to stimulate the synthesis and accumulation of ECM, leading to fibrosis and hypertrophy in renal cells under diabetic conditions (23, 60, 129, 165). Smad2/Smad3 transcription factors are major effectors of TGF-β1 actions (122, 168) and regulate the expression of TGF-β1 target fibrotic genes (118, 142). These transcription factors are regulated via TGF-β1-mediated activation of key signaling kinases, such as mitogen-activated protein kinases (24, 49) and Akt kinase [by inhibiting phosphatase and tensin homolog (PTEN)] (69, 94).

Other than these classic factors implicated in DN, emerging evidence suggests that miRNAs can play significant roles in the early stages of DN and ESRD (64, 66, 141). Notably, severe glomerular and tubular injury, proteinuria, and renal failure were observed in mice with podocyte-specific deletion of Dicer, an essential and important nuclease involved in miRNA biogenesis (production), suggesting the involvement of miRNAs in kidney dysfunction (48, 52, 130). However, because this strategy inhibits several miRNAs, the functions of specific miRNAs in renal failure were not clear from these studies. Early comprehensive studies showed that specific miRNAs are enriched in the kidney compared with other organs (135), and furthermore different miRNA expression patterns were observed in the renal cortex vs. medulla, suggesting their cell type- and tissue-specific functions (138). Several miRNAs (Table 1) have now been shown to play critical roles in DN because they can target key transcription factors, fibrotic, inflammatory, cell cycle, or other genes associated with DN. In the first demonstration of a functional role for a kidney miRNA in DN, miR-192 was found to be upregulated by TGF-β1 and in the cortex of diabetic mice. miR-192 could upregulate the key ECM and fibrotic gene collagen, type I, α2 (Col1a2) in mesangial cells (MC) by targeting the E-box repressors Zeb1 and Zeb2 (which control the expression of TGF-β1-induced collagen) (70). Interestingly, miR-192 was found to upregulate other miRNAs such as miR-216a/miR-217 and miR-200b/c through targeting Zeb1 and Zeb2, initiating amplifying circuits to further augment fibrotic gene expression in DN (61, 67). Both miR-216a and miR-217 were shown to target PTEN, leading to the activation of Akt kinase in mouse MC treated with TGF-β1, which in turn could enhance cellular hypertrophy (67). miR-200b/c in turn were shown to upregulate collagen expression and initiate the autoregulation of TGF-β1 in MMC by targeting Zeb1 (61). Furthermore, other studies showed that miR-200b/c also activate Akt by targeting FOG2, an inhibitor of phosphoinositide 3-kinase (PI3K) (110). It is interesting to determine the mechanisms by which specific miRNAs are transcribed and regulated under diabetic conditions. Interestingly, TGF-β1-induced regulation of the miR-192 promoter in MMC involved not only Smads, but also Ets-1 and p53 transcription factors, and chromatin remodeling through histone acetylation by the histone acetyltransferase p300, which is activated by Akt kinase (30, 63, 66). miR-192 promoter was also regulated by hepatocyte nuclear factor-1 (HNF-1) in other cell types (54, 66). Multiple other studies have reported these miRNAs (miR-192, -200 family, 216a, -217) to be induced in renal cells treated with HG or TGF-β1, or in animal models of kidney injury (26, 50, 101, 134, 157). Moreover, diabetic miR-192 knockout mice or diabetic mice treated with a oligonucleotide miR-192 inhibitor were protected from key features of DN (30, 119), suggesting miR-192 can serve as a major upstream miRNA regulator of genes and factors involved in the early stage of DN (26, 30, 61, 63, 6668, 70, 110).

Table 1.

Concise list of miRNAs involved in the pathogenesis of diabetic nephropathy and acute kidney injury

Disease miRNA Targets Pathways Function Selected Reference(s)
DN miR-192 δEF1, SIP1, ZEB1/2 TGF-β, AKT Pathogenic 61, 67, 70
DN miR-216a/217 PTEN, YBX1 TGF-β, AKT Pathogenic 67
DN miR-200b/c ZEB1, FOG2 TGF-β, AKT Pathogenic 61, 110
DN miR-21 PTEN, PRAS40, SMAD7, TIMP1/3 TGF-β, AKT Pathogenic 32, 153, 169, 170
DN miR-25 NOX4 Oxidative stress Protective 41
DN miR-377 MnSOD, PAK Extracellular matrix Pathogenic 156
DN miR-29 Collagens Extracellular matrix Protective 22, 34, 148
DN miR-29c SPRY1 Rho kinase Pathogenic 88
DN Let-7b TGFBR1 Collagens TGF-β Extracellular matrix Protective 15, 109, 146
DN miR-130b TGFBR1 TGF-β Protective 17
DN miR-93 VEGFA VEGF Protective 87
DN miR-26a CTGF TGF-β/CTGF Protective 31, 71
AKI miR-24 H2AX, HO-1 Oxidative stress Pathogenic 89
AKI miR-494 ATF3 ER stress Pathogenic 78
AKI miR-687 PTEN Hypoxia, cell cycle Pathogenic 11
AKI miR-150 IGF1R IGF Pathogenic 121
AKI miR-126 PI3KR2 PI3K/AKT Protective 14
AKI miR-127 KIF3B Cell trafficking Protective 2

miRNA, microRNA; DN, diabetic nephropathy; AKI, acute kidney injury; TGF, transforming growth factor. See the text for additional definitions.

Another miRNA that has been widely studied and implicated in several cell and animal models of DN and kidney injury is miR-21 because its targets (which include PTEN) are related to fibrosis, Akt activation, hypertrophy, and apoptosis (32, 153, 170). Recently miR-21 was shown to promote fibrosis by targeting metabolic pathways. Relative to wild-type or untreated mice, miR-21 knockout mice or mice treated with anti-miR-21 oligonucleotides showed less severe interstitial fibrosis in response to renal injury (20). On the other hand, miR-21 was downregulated in db/db mice, and its overexpression inhibited MC proliferation (169). More severe renal phenotypes were detected in miR-21 knockout mice crossed with TGF-β1 transgenic mice relative to TGF-β1 transgenic mice (77). Interestingly some miRNAs like the miR-29 family members can directly regulate ECM accumulation because these miRNAs target multiple collagens and hence a decrease in their levels can augment collagen deposition in the kidney or in renal cells treated with TGF-β1 or HG (22, 34, 73, 148). Importantly, miR-29b overexpression was found to significantly alleviate DN in db/db mice (22). Interestingly, an antidiabetic drug, linagliptin, a dipeptidyl peptidase-4 (DPP-4) inhibitor, was shown to upregulated miR-29 and prevent fibrosis in a mouse model of DN (59). On the other hand, in another study miR-29c was found to be upregulated under diabetic conditions and activated Rho kinase by targeting Spry-1, leading to ECM accumulation and podocyte apoptosis (88). Other miRNAs have also been studied in DN. MiR-377 upregulated by HG or TGF-β1 in MC induced fibronectin expression and oxidant stress by targeting manganese superoxide dismutase and p21-activated kinase (156). Let-7 family members known as tumor suppressors (55, 97, 125) were also downregulated by TGF-β1 in renal cells, and this could promote fibrosis through upregulation of the let-7 targets TGF-β1 receptor type-1 (TGF-β1R1) and collagens (15, 109, 146). Mechanistically, it was interesting that let-7 family members downregulated by TGF-β1 induced upregulation of lin28b (negative regulator of let-7 processing) through Smad2/3 (109). The let-7-TGF-β1R1 connection illustrates another signal-amplifying loop involving miRNAs that can accelerate DN (65, 66). Interestingly, the downregulation of let-7a-3 in DN was associated with promoter DNA hypermethylation, suggesting an epigenetic regulatory mechanism (114). TGF-β1-induced decrease of miR-130b was shown to upregulate its target, TGF-β1R1, in MC through mechanisms involving a decrease in Ybx1/NFYC targeted by miR-216a, thus revealing another miRNA-mediated amplifying cascade (17). miR-135a, which promotes ECM accumulation by targeting a transient receptor potential cation channel, subfamily C, member 1 (TRPC1), was enriched in serum and renal tissue from patients with DN and db/db mice (50). miR-30 family members were decreased in accelerated DN, and this was related to the upregulation of their target, connective tissue growth factor (CTGF) (162). miR-22 regulates BMP-7 and BMP-6 and further increases TGF-β1 signaling in mouse models of kidney fibrosis (86). miR-93 was identified as a key miRNA downregulated in podocytes and renal microvascular endothelial cells treated with HG as well as in glomeruli of diabetic db/db mice, and this enhanced angiogenesis via an increase in its target, VEGF-A (87). miR-26a was downregulated in DN patients and also by TGF-β1 in cultured podocytes, leading to an increase in its target, CTGF (71). Interestingly, miR-26a was also upregulated by HG in MC and could activate mTORC1, enhance hypertrophy, and ECM accumulation (31).

Oxidant stress is another important factor in the pathogenesis of DN, and several miRNAs that modulate oxidant stress have been identified (6, 39, 41, 102, 127, 171). The balance of pro-oxidant miRNAs and antioxidant miRNAs may be a key to control renal oxidant stress and DN (62).

Notably, distinct effects of key miRNAs in renal cells have been reported. For example, in some other models of more severe or late stages of DN and in some studies of cultured proximal tubular epithelial cells treated with HG or TGF-β1, decreases in miR-192 were reported (74, 145), although as noted earlier miR-192 was upregulated in renal cells treated with TGF-β1 or HG and many animal models of earlier DN in several studies (26, 30, 61, 63, 6668, 70, 110). miR-26a was downregulated by TGF-β1 in cultured podocytes (71) although it was upregulated by HG in cultured MC (31). Cell type-specific effects of miRNAs and differences in the animal models studied may be the reason. The cell type-specific response of miR-192 to TGF-β1 can also be explained by the cellular status of transcription factors such as p53, Ets-1, or Smads, since TGF-β1-induced miR-192 expression was abolished in MC from p53−/−, Ets-1−/−, or Smad3−/− mice (26, 30, 63). miR-200 family members were increased related to ECM accumulation and Akt activation in primary MMC (61, 110), although miR-200a was downregulated in TGF-β1-treated proximal tubular epithelial cells also related to fibrosis through TGF-β2 (147). Importantly, the effects of miRNAs such as miR-200 can vary in primary epithelial cells vs. cancer cells (66). Furthermore, it is likely that miRNA levels are lower in renal tissues obtained from patients in late stages of DN relative to early (74), due to poor quality of RNA arising from tissue necrosis and apoptotic cells. Thus it is worthwhile to profile miRNAs in models of early DN (including animal models), rather than late stage, as such miRNAs can be good targets to prevent progression to overt renal failure (30, 65, 66, 119). This is illustrated by data showing that inhibition of miR-192, which is highly expressed in mouse models of early DN (63, 64, 70) but decreased in biopsies from patients in late stages of DN (74), prevented key features of DN in animal models (30, 119).

An area that is showing great interest and promise is in harnessing the potential of miRNAs as novel biomarkers. Early diagnosis of DN is of tremendous value to prevent progression to renal failure and dialysis. Since there have been significant advances in sensitive and precise detection of miRNAs in human biofluids such as urine and plasma, miRNAs are emerging as promising candidate noninvasive biomarkers for human diseases (36, 38, 139, 154). Comprehensive profiles of miRNAs in urine, urinary sediment, and serum from patients in specific stages of DN, fibrosis, renal function decline (GFR), albuminuria, or rapid progression to ESRD have been reported (5, 8, 13, 16, 33, 50, 51, 92, 93, 106, 115, 136, 151, 152, 166). Because molecular mechanisms underlying DN may vary from patient to patient, individual miRNA profiles could be used as a key factor for personalized medicine and treatment (65). Another area of active research is miRNA-based therapy, and several delivery approaches in animal models of DN have been attempted to control miRNA expression in vivo (33, 64, 66). Chemically modified stable nuclease-resistant oligonucleotides have been developed that might be used in future for patients. Antisense miRNAs modified with Locked nucleic acid (LNA) are relatively popular due to their superior targeting efficiency (67, 75, 119) and also being evaluated in some clinical trials (82, 144). Targeting several miRNAs by LNA-modified or 2′-O-methyl antisense oligonucleotides has been successful to reduce specific miRNAs and inhibit fibrosis and hypertrophy in mouse models of DN and fibrosis (20, 30, 50, 61, 67, 88, 119, 170). It is anticipated that more sensitive and precise methods to monitor miRNAs in human biofluids will be established, while success in preventing DN in animal models with chemically-modified oligonucleotide inhibitors of specific miRNAs provides hope for clinical translation to human DN in the future.

Acute Kidney Injury

Acute kidney injury (AKI) is characterized by a rapid decline in renal function due to ischemic or toxic damage in renal tubular cells (10). Mortality rates for AKI are extremely high in part due to the lack of sensitive diagnostic markers and an absence of therapeutic options (10). Importantly, it has been recognized that AKI is a key contributing factor in the development of chronic kidney disease (CKD) (21). Even mild AKI episodes can lead to CKD and an increased long-term risk of developing ESRD (21).

Recent studies have revealed the critical role played by miRNAs in modulating multiple cellular and molecular pathways that contribute to AKI (159). In one of the initial studies (158) examining the functional role of miRNAs in AKI, it was found that global miRNA depletion by conditional Dicer ablation in renal tubular cells provides significant protection from ischemic AKI. This study established the pathogenic role of Dicer and associated miRNAs in ischemic AKI. Subsequent studies (159) have now unveiled both protective and pathogenic roles of specific miRNAs in the development of AKI. Based on these studies, several miRNAs (Table 1) have been postulated to have diagnostic utility, while miRNA-based therapeutic strategies have also been suggested for AKI.

Renal tubular cell death is the major contributing factor in the development of AKI (83). Multiple miRNAs have been shown to contribute to renal tubular cell apoptosis. The apoptosis-associated miR-24 is upregulated in the murine kidneys after ischemia-reperfusion (I/R) injury and in patients after kidney transplantation (89). Importantly, silencing of miR-24 in mouse models of I/R injury resulted in significant renoprotection. The apoptotic role of miR-24 was found to be mediated through direct regulation of two target genes, namely, H2A histone family member X and heme oxygenase 1 (89). These results indicate that miR-24 inhibition may be a promising therapeutic option in the treatment of patients with ischemic AKI. miR-494 is another cell death-associated miRNA, which plays a pathogenic role during AKI (78). Initial studies found that urinary miR-494 levels were many-fold higher in patients with AKI than normal controls. In mouse models, renal overexpression of miR-494 led to increased renal damage, which was attributed to reduced levels of its target gene, activating transcription factor 3 (ATF3) (78). Similar to miR-24 and miR-494, miR-687 has recently (11) been proposed to contribute to I/R-mediated AKI. This study (11) identified a novel hypoxia-inducible factor HIF-1/miR-687/PTEN signaling pathway, where miR-687 antagonism preserved PTEN expression, blocking cell cycle activation and preventing renal apoptosis, resulting in significant protection in mouse models of AKI. In a novel myocardial infarction-induced AKI mouse model, miR-150 deficiency also suppressed AKI, which was associated with suppression of inflammation and interstitial cell apoptosis (121). Interestingly, preischemic conditioning protected mice from AKI, and these effects were not observed in miR-21 knockout mice, suggesting that miR-21 plays a protective role during AKI(164).

Along with miRNAs that contribute to AKI, renoprotective miRNAs have also been identified. Bijkerk et al. (14) showed that overexpression of miR-126 in the hematopoietic compartment can protect the kidney via preservation of microvascular integrity during ischemic renal injury and supports recovery by promoting vasculogenic progenitor cell mobilization. In another study (2), miR-127 was identified as a renoprotective miRNA during ischemic renal injury. Ischemic induction of miR-127 was found to be mediated by HIF-1α stabilization, and functional studies showed that miR-127 is involved in cell matrix and cell-cell adhesion maintenance. Similarly, the p53-inducible miR-34a may play a cytoprotective role during cisplatin-induced renal tubular cell death (12).

Besides their roles in pathogenesis or protection from AKI, studies (9, 76, 113, 160) on miRNA expression profiles have illustrated the usefulness of miRNAs as biomarkers for diagnosis of AKI. Although, extensive human studies are still required, initial steps have been taken to demonstrate the potential utility of miRNAs, especially in the urine, for assessing AKI. Multiple studies (90, 96, 104, 113, 124, 155, 159, 160) in humans and animal models have identified specific miRNAs as potential diagnostic markers for the early detection of AKI. Such approaches would be of tremendous clinical value to prevent progression from AKI to renal failure.

IgA Nephropathy

IgA nephropathy is a common form of glomerulonephritis that can lead to ESRD (163). The major hallmark of IgA nephropathy is glomerular deposition of IgA immune complexes, causing activation of mesangial and inflammatory cells, resulting in renal injury (99). miRNAs have been implicated in the pathogenesis of IgA nephropathy and have received significant attention as potential diagnostic and prognostic markers (137, 167). Interestingly, aberrant O-glycosylation in the hinge region of IgA1 is a common feature of IgA nephropathy (137). In an important study (128), it was found that this aberrant O-glycosylation of IgA1 is due to increased expression of miR-148b, which targets the glycosylation enzyme, β1,3-galactosyltransferase 1 (C1GALT1). This study (128) provides a direct link between the overexpression of miR-148 and the aberrant glycosylation of IgA1, which plays a pathogenic role in the early phase of IgA nephropathy. The miR-29 family which targets several extracellular matrix genes has also been implicated in the pathogenesis of IgA nephropathy (137). In particular the HIF-1α-dependent miR-29c has been suggested to play an anti-fibrotic role by down-regulating collagen type II α1 (COL2A1) protein, and tropomyosin 1α (TPM1) proteins (37). Global analysis of renal miRNA expression has also identified several miRNAs related to immunological and pathological changes that might contribute to the development or progression of IgA nephropathy (137). Additionally, urinary levels of several miRNAs are significantly altered in IgA nephropathy patients and have the potential to be used as biomarkers for diagnostic purposes (58, 104, 137).

Polycystic Kidney Disease

Polycystic kidney disease (PKD) is an inherited disorder characterized by development of clusters of cysts within the kidneys (47). The inherited mutations are frequently observed within specific genes, including PKD1, PKD2, and PKHD1, which contribute to abnormal proliferation resulting in cyst formation (19). Recent studies with global miRNA profiling have shown that the miRNA expression pattern is significantly altered in PKD models, suggesting that miRNAs may act as disease modulators downstream of PKD mutations (25, 108). In an interesting study (112), it was shown that the miR-17∼92 cluster is upregulated in mouse models of PKD. Moreover, transgenic overexpression of miR-17∼92 in kidneys produced cysts in normal mice, while inactivation of miR-17∼92 in mouse models of PKD retarded kidney cyst growth and provided significant survival benefits. Mechanistic studies showed that the miR-17∼92 cluster promotes aberrant renal cell proliferation through repression of several PKD genes (112).

Other studies have also shown that the PKD genes are miRNA targets, providing a direct link between PKD gene expression, miRNAs, and cystogenesis (116, 141). For example, miR-17 has been shown to target PKD2 and overexpression of miR-17 can promote aberrant cellular proliferation in cell culture models (133). Moreover, transgenic mice expressing miRNAs targeting PKD1 develop several pathophysiological features of PKD (149). Interestingly, knockout mice of bicaudal C (Bicc1), an evolutionarily conserved RNA-binding protein, develop a PKD phenotype (140). Mechanistic studies showed that Bicc1 antagonizes the function of miR-17 and stabilizes PKD2 mRNA. This was substantiated in Xenopus models, where the renal defects of Bicc1 knockdown were rescued by reducing miR-17 activity (140). These studies (112, 117, 133, 140) have provided solid proof-of-principle evidence and a strong rationale for targeting miR-17 as a therapeutic strategy in the treatment of PKD.

Autosomal recessive PKD (ARPKD), a subtype of PKD, is generally caused by mutation in the polycystic kidney and hepatic disease 1 (PKHD1) gene (46). PKHD1 was found to be a direct target of miR-365-1 (35). Polycystic liver disease, a frequent feature of PKD, is associated with a reduction in miR-15a expression (79). Loss or reduction in miR-15a expression could directly contribute to aberrant cell cycle progression through increased expression of cell cycle-regulatory genes like cdc25a (79).

Other Renal Diseases and Conditions

Along with the above-mentioned renal diseases, miRNAs have also been studied in lupus nephritis, acute allograft rejection, hypertensive nephropathy, and other disorders (141, 159). Altered expression of several miRNAs has been demonstrated in lupus nephritis (131, 132, 141). These differentially expressed miRNAs include miR-638, miR-663, miR198, miR-155, and miR-146a (91, 141). Similarly, acute allograft rejection, a frequent complication of renal transplantation, is associated with alterations in miRNA expression (4, 84, 95, 143, 160). Global miRNA profiling of human hypertensive nephrosclerosis biopsies also revealed a number of differentially expressed miRNAs, including miR-200a, miR-200b, miR-141, miR-429, and miR-192 (150). Similar profiling has been performed in Dahl salt-sensitive rats (103). miR-29b is upregulated in renal medullary tissues in Dahl salt-sensitive rats fed a high-salt diet, and miR-29b overexpression in cultured cells can suppress collagen expression (85). These findings demonstrate a negative regulatory role of miR-29b in collagen and extracellular matrix accumulation in hypertensive renal injury. Interestingly, recent work has implicated miR-217 in mediating the protective effects of the dopamine D2 receptor on fibrosis in renal tubular cells (45). Additionally, miR-192 and mir-324-3p may play a role in hypertensive nephropathy (150). In another important study, miR-382 was found to target kallikrein 5 and contributes to the development of renal inner medullary interstitial fibrosis (72).

miRNAs have also been implicated in focal segmental glomerulosclerosis (FSGS). Higher expression of miR-193a was observed in isolated glomeruli from individuals with FSGS compared with normal subjects, and transgenic overexpression of miR-193a in mice induced FSGS and podocyte foot process effacement. This was mechanistically attributed to miR-193a-mediated targeting of Wilms' tumor protein (WT1), a critical regulator of podocyte differentiation and homeostasis, and was associated with downregulation of key WT1 target genes, including podocalyxin and nephrin (42). Increased expression of miR-21 was also observed in Alport nephropathy, a genetic disorder characterized by chronic kidney disease, such as glomerulonephritis. Notably, in this study it was demonstrated that inhibition of miR-21 by antisense oligonucleotides in a mouse model prevented typical features of this disease, including glomerulosclerosis, interstitial fibrosis, tubular injury, and inflammation (43). It is important to note that despite the evidence of differential expression of various miRNAs and their potential functions in renal physiology or pathology, the functional significance in many cases remains unknown and is expected to remain an area of active investigation.

Conclusions and Future Perspectives

Significant progress has been made in our understanding of how miRNAs regulate normal and abnormal kidney function. Strong experimental data in animal models have emerged showing the role of specific miRNAs in the development and progression of renal diseases. Moreover, it has become possible to target miRNAs in vivo (to inhibit or restore miRNA expression), which provides hope for the future development of renal miRNA therapeutics. Furthermore, miRNAs have significant potential as novel diagnostic and prognostic biomarkers for kidney diseases. However, several challenges still remain for translating animal studies to the clinic. Some of these challenges include targeting of miRNA-based therapeutics to specific renal cells and the possibility that more than one miRNA may have to be targeted to have significant clinical benefits. Furthermore, as discussed earlier, miRNAs can contribute to the biological processes affecting kidney functions via direct as well as indirect effects. Added to this complexity of miRNA-mediated actions, one miRNA can target multiple genes and, furthermore, since miRNAs can have cell-specific effects, this poses significant challenges especially in a heterogeneous organ like the kidney. In terms of their clinical diagnostic potential, there is a need for intensive clinical studies in appropriate cohorts with patients from various stages of the disease to identify which miRNA(s) could be used as biomarkers of disease progression. Several studies have identified differentially expressed miRNAs in the blood, urine, and kidney, raising the possibility of developing miRNAs as diagnostic markers. However, these studies have one or more limitations, including insufficient sample size, single time points, lack of appropriate controls, insufficient information on the effect of other comorbidities, and lack of secondary validations. On the other hand, the uniqueness of the miRNAs differentially expressed in the diseases of interest and the cell type- or tissue-specific expression may render them as valuable and specific biomarkers. Notwithstanding these challenges, pitfalls, and promises, understanding the role of miRNAs in renal diseases has a significant potential to lead to the development of a novel class of diagnostic and therapeutic tools for the early detection and treatment of renal diseases.

GRANTS

The authors gratefully acknowledge funding from National Institute of Health Grants R01 DK081705, R01 DK058191, and R01 DK065073 (to R. Natarajan) and a Juvenile Diabetes Research Foundation (JDRF) postdoctoral fellowship (to K. Bhatt).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

Author contributions: K.B., M.K., and R.N. drafted manuscript; K.B., M.K., and R.N. edited and revised manuscript; R.N. approved final version of manuscript.

REFERENCES

  • 1.Abboud HE. Role of platelet-derived growth factor in renal injury. Annu Rev Physiol 57: 297–309, 1995. [DOI] [PubMed] [Google Scholar]
  • 2.Aguado-Fraile E, Ramos E, Saenz-Morales D, Conde E, Blanco-Sanchez I, Stamatakis K, del Peso L, Cuppen E, Brune B, Bermejo ML. 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]
  • 3.Ameres SL, Zamore PD. Diversifying microRNA sequence and function. Nat Rev Mol Cell Biol 14: 475–488, 2013. [DOI] [PubMed] [Google Scholar]
  • 4.Anglicheau D, Sharma VK, Ding R, Hummel A, Snopkowski C, Dadhania D, Seshan SV, Suthanthiran M. MicroRNA expression profiles predictive of human renal allograft status. Proc Natl Acad Sci USA 106: 5330–5335, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Argyropoulos C, Wang K, McClarty S, Huang D, Bernardo J, Ellis D, Orchard T, Galas D, Johnson J. Urinary microRNA profiling in the nephropathy of type 1 diabetes. PLoS One 8: e54662, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Babelova A, Avaniadi D, Jung O, Fork C, Beckmann J, Kosowski J, Weissmann N, Anilkumar N, Shah AM, Schaefer L, Schroder K, Brandes RP. Role of Nox4 in murine models of kidney disease. Free Radic Biol Med 53: 842–853, 2012. [DOI] [PubMed] [Google Scholar]
  • 7.Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 136: 215–233, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Barutta F, Tricarico M, Corbelli A, Annaratone L, Pinach S, Grimaldi S, Bruno G, Cimino D, Taverna D, Deregibus MC, Rastaldi MP, Perin PC, Gruden G. Urinary exosomal microRNAs in incipient diabetic nephropathy. PLoS One 8: e73798, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bellinger MA, Bean JS, Rader MA, Heinz-Taheny KM, Nunes JS, Haas JV, Michael LF, Rekhter MD. 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]
  • 10.Bellomo R, Kellum JA, Ronco C. Acute kidney injury. Lancet 380: 756–766, 2012. [DOI] [PubMed] [Google Scholar]
  • 11.Bhatt K, Wei Q, Pabla N, Dong G, Mi QS, Liang M, Mei C, Dong Z. MicroRNA-687 induced by hypoxia-inducible factor-1 targets phosphatase and tensin homolog in renal ischemia-reperfusion injury. J Am Soc Nephrol 26: 1588–1596, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bhatt K, Zhou L, Mi QS, Huang S, She JX, Dong Z. 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]
  • 13.Bijkerk R, Duijs JMGJ, Khairoun M, ter Horst CJH, van der Pol P, Mallat MJ, Rotmans JI, de Vries APJ, de Koning EJ, de Fijter JW, Rabelink TJ, van Zonneveld AJ, Reinders MEJ. Circulating microRNAs associate with diabetic nephropathy and systemic microvascular damage and normalize after simultaneous pancreas-kidney transplantation. Am J Transplant 15: 1081–1090, 2015. [DOI] [PubMed] [Google Scholar]
  • 14.Bijkerk R, van Solingen C, de Boer HC, van der Pol P, Khairoun M, de Bruin RG, van Oeveren-Rietdijk AM, Lievers E, Schlagwein N, van Gijlswijk DJ, Roeten MK, Neshati Z, de Vries AA, Rodijk M, Pike-Overzet K, van den Berg YW, van der Veer EP, Versteeg HH, Reinders ME, Staal FJ, van Kooten C, Rabelink TJ, van Zonneveld AJ. 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]
  • 15.Brennan EP, Nolan KA, Borgeson E, Gough OS, McEvoy CM, Docherty NG, Higgins DF, Murphy M, Sadlier DM, Ali-Shah ST, Guiry PJ, Savage DA, Maxwell AP, Martin F, Godson C, Consortium G. Lipoxins attenuate renal fibrosis by inducing let-7c and suppressing TGFbetaR1. J Am Soc Nephrol 24: 627–637, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cai X, Xia Z, Zhang C, Luo Y, Gao Y, Fan Z, Liu M, Zhang Y. Serum microRNAs levels in primary focal segmental glomerulosclerosis. Pediatr Nephrol 28: 1797–1801, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Castro NE, Kato M, Park JT, Natarajan R. Transforming growth factor beta1 (TGF-beta1) enhances expression of profibrotic genes through a novel signaling cascade and microRNAs in renal mesangial cells. J Biol Chem 289: 29001–29013, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Cech TR, Steitz JA. The noncoding RNA revolution-trashing old rules to forge new ones. Cell 157: 77–94, 2014. [DOI] [PubMed] [Google Scholar]
  • 19.Chapin HC, Caplan MJ. The cell biology of polycystic kidney disease. J Cell Biol 191: 701–710, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chau BN, Xin C, Hartner J, Ren S, Castano AP, Linn G, Li J, Tran PT, Kaimal V, Huang X, Chang AN, Li S, Kalra A, Grafals M, Portilla D, MacKenna DA, Orkin SH, Duffield JS. MicroRNA-21 promotes fibrosis of the kidney by silencing metabolic pathways. Sci Transl Med 4: 121ra118, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.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]
  • 22.Chen HY, Zhong X, Huang XR, Meng XM, You Y, Chung AC, Lan HY. MicroRNA-29b inhibits diabetic nephropathy in db/db mice. Mol Ther 22: 842–853, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chen S, Jim B, Ziyadeh FN. Diabetic nephropathy and transforming growth factor-beta: transforming our view of glomerulosclerosis and fibrosis build-up. Semin Nephrol 23: 532–543, 2003. [DOI] [PubMed] [Google Scholar]
  • 24.Chin BY, Mohsenin A, Li SX, Choi AM, Choi ME. Stimulation of pro-α1(I) collagen by TGF-β1 in mesangial cells: role of the p38 MAPK pathway. Am J Physiol Renal Physiol 280: F495–F504, 2001. [DOI] [PubMed] [Google Scholar]
  • 25.Chu AS, Friedman JR. A role for microRNA in cystic liver and kidney diseases. J Clin Invest 118: 3585–3587, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Chung AC, Huang XR, Meng X, Lan HY. miR-192 mediates TGF-beta/Smad3-driven renal fibrosis. J Am Soc Nephrol 21: 1317–1325, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Colagiuri S, Cull CA, Holman RR. Are lower fasting plasma glucose levels at diagnosis of type 2 diabetes associated with improved outcomes?: UK Prospective Diabetes Study 61. Diabetes Care 25: 1410–1417, 2002. [DOI] [PubMed] [Google Scholar]
  • 28.de Boer IH, Sun W, Cleary PA, Lachin JM, Molitch ME, Steffes MW, Zinman B, Group DER. Intensive diabetes therapy and glomerular filtration rate in type 1 diabetes. N Engl J Med 365: 2366–2376, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.de Planell-Saguer M, Rodicio MC. Analytical aspects of microRNA in diagnostics: a review. Anal Chim Acta 699: 134–152, 2011. [DOI] [PubMed] [Google Scholar]
  • 30.Deshpande SD, Putta S, Wang M, Lai JY, Bitzer M, Nelson RG, Lanting LL, Kato M, Natarajan R. Transforming growth factor-beta-induced cross talk between p53 and a microRNA in the pathogenesis of diabetic nephropathy. Diabetes 62: 3151–3162, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Dey N, Bera A, Das F, Ghosh-Choudhury N, Kasinath BS, Choudhury GG. High glucose enhances microRNA-26a to activate mTORC1 for mesangial cell hypertrophy and matrix protein expression. Cell Signal 27: 1276–1285, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Dey N, Das F, Mariappan MM, Mandal CC, Ghosh-Choudhury N, Kasinath BS, Choudhury GG. MicroRNA-21 orchestrates high glucose-induced signals to TOR complex 1, resulting in renal cell pathology in diabetes. J Biol Chem 286: 25586–25603, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.DiStefano JK, Taila M, Alvarez ML. Emerging roles for miRNAs in the development, diagnosis, and treatment of diabetic nephropathy. Curr Diabetes Rep 13: 582–591, 2013. [DOI] [PubMed] [Google Scholar]
  • 34.Du B, Ma LM, Huang MB, Zhou H, Huang HL, Shao P, Chen YQ, Qu LH. High glucose down-regulates miR-29a to increase collagen IV production in HK-2 cells. FEBS Lett 584: 811–816, 2010. [DOI] [PubMed] [Google Scholar]
  • 35.Duan J, Huang H, Lv X, Wang H, Tang Z, Sun H, Li Q, Ai J, Tan R, Liu Y, Chen M, Duan W, Wei Y, Zhou Q. PKHD1 post-transcriptionally modulated by miR-365-1 inhibits cell-cell adhesion. Cell Biochem Funct 30: 382–389, 2012. [DOI] [PubMed] [Google Scholar]
  • 36.Fabbri M. miRNAs as molecular biomarkers of cancer. Expert Rev Mol Diagn 10: 435–444, 2010. [DOI] [PubMed] [Google Scholar]
  • 37.Fang Y, Yu X, Liu Y, Kriegel AJ, Heng Y, Xu X, Liang M, Ding X. miR-29c is downregulated in renal interstitial fibrosis in humans and rats and restored by HIF-α activation. Am J Physiol Renal Physiol 304: F1274–F1282, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Farazi TA, Spitzer JI, Morozov P, Tuschl T. miRNAs in human cancer. J Pathol 223: 102–115, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Feng B, Chen S, McArthur K, Wu Y, Sen S, Ding Q, Feldman RD, Chakrabarti S. miR-146a-Mediated extracellular matrix protein production in chronic diabetes complications. Diabetes 60: 2975–2984, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Fineberg D, Jandeleit-Dahm KA, Cooper ME. Diabetic nephropathy: diagnosis and treatment. Nat Rev Endocrinol 9: 713–723, 2013. [DOI] [PubMed] [Google Scholar]
  • 41.Fu Y, Zhang Y, Wang Z, Wang L, Wei X, Zhang B, Wen Z, Fang H, Pang Q, Yi F. Regulation of NADPH oxidase activity is associated with miRNA-25-mediated NOX4 expression in experimental diabetic nephropathy. Am J Nephrol 32: 581–589, 2010. [DOI] [PubMed] [Google Scholar]
  • 42.Gebeshuber CA, Kornauth C, Dong L, Sierig R, Seibler J, Reiss M, Tauber S, Bilban M, Wang S, Kain R, Bohmig GA, Moeller MJ, Grone HJ, Englert C, Martinez J, Kerjaschki D. Focal segmental glomerulosclerosis is induced by microRNA-193a and its downregulation of WT1. Nat Med 19: 481–487, 2013. [DOI] [PubMed] [Google Scholar]
  • 43.Gomez IG, MacKenna DA, Johnson BG, Kaimal V, Roach AM, Ren S, Nakagawa N, Xin C, Newitt R, Pandya S, Xia TH, Liu X, Borza DB, Grafals M, Shankland SJ, Himmelfarb J, Portilla D, Liu S, Chau BN, Duffield JS. Anti-microRNA-21 oligonucleotides prevent Alport nephropathy progression by stimulating metabolic pathways. J Clin Invest 125: 141–156, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol 15: 509–524, 2014. [DOI] [PubMed] [Google Scholar]
  • 45.Han F, Konkalmatt P, Chen J, Gildea J, Felder RA, Jose PA, Armando I. MiR-217 mediates the protective effects of the dopamine D2 receptor on fibrosis in human renal proximal tubule cells. Hypertension 65: 1118–1125, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Harris PC, Rossetti S. Molecular diagnostics for autosomal dominant polycystic kidney disease. Nat Rev Nephrol 6: 197–206, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Harris PC, Torres VE. Polycystic kidney disease. Annu Rev Med 60: 321–337, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Harvey SJ, Jarad G, Cunningham J, Goldberg S, Schermer B, Harfe BD, McManus MT, Benzing T, Miner JH. Podocyte-specific deletion of dicer alters cytoskeletal dynamics and causes glomerular disease. J Am Soc Nephrol 19: 2150–2158, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hayashida T, Poncelet AC, Hubchak SC, Schnaper HW. TGF-beta1 activates MAP kinase in human mesangial cells: a possible role in collagen expression. Kidney Int 56: 1710–1720, 1999. [DOI] [PubMed] [Google Scholar]
  • 50.He F, Peng F, Xia X, Zhao C, Luo Q, Guan W, Li Z, Yu X, Huang F. MiR-135a promotes renal fibrosis in diabetic nephropathy by regulating TRPC1. Diabetologia 57: 1726–1736, 2014. [DOI] [PubMed] [Google Scholar]
  • 51.Higuchi C, Nakatsuka A, Eguchi J, Teshigawara S, Kanzaki M, Katayama A, Yamaguchi S, Takahashi N, Murakami K, Ogawa D, Sasaki S, Makino H, Wada J. Identification of circulating miR-101, miR-375 and miR-802 as biomarkers for type 2 diabetes. Metab Clin Exp 64: 489–497, 2015. [DOI] [PubMed] [Google Scholar]
  • 52.Ho J, Ng KH, Rosen S, Dostal A, Gregory RI, Kreidberg JA. Podocyte-specific loss of functional microRNAs leads to rapid glomerular and tubular injury. J Am Soc Nephrol 19: 2069–2075, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, van der Meer AJ, Patick AK, Chen A, Zhou Y, Persson R, King BD, Kauppinen S, Levin AA, Hodges MR. Treatment of HCV infection by targeting microRNA. N Engl J Med 368: 1685–1694, 2013. [DOI] [PubMed] [Google Scholar]
  • 54.Jenkins RH, Martin J, Phillips AO, Bowen T, Fraser DJ. Transforming growth factor beta1 represses proximal tubular cell microRNA-192 expression through decreased hepatocyte nuclear factor DNA binding. Biochem J 443: 407–416, 2012. [DOI] [PubMed] [Google Scholar]
  • 55.Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, Labourier E, Reinert KL, Brown D, Slack FJ. RAS is regulated by the let-7 microRNA family. Cell 120: 635–647, 2005. [DOI] [PubMed] [Google Scholar]
  • 56.Jonas S, Izaurralde E. Towards a molecular understanding of microRNA-mediated gene silencing. Nat Rev Genet 16: 421–433, 2015. [DOI] [PubMed] [Google Scholar]
  • 57.Jones CA, Krolewski AS, Rogus J, Xue JL, Collins A, Warram JH. Epidemic of end-stage renal disease in people with diabetes in the United States population: do we know the cause? Kidney Int 67: 1684–1691, 2005. [DOI] [PubMed] [Google Scholar]
  • 58.Julian BA, Wittke S, Haubitz M, Zurbig P, Schiffer E, McGuire BM, Wyatt RJ, Novak J. Urinary biomarkers of IgA nephropathy and other IgA-associated renal diseases. World J Urol 25: 467–476, 2007. [DOI] [PubMed] [Google Scholar]
  • 59.Kanasaki K, Shi S, Kanasaki M, He J, Nagai T, Nakamura Y, Ishigaki Y, Kitada M, Srivastava SP, Koya D. Linagliptin-mediated DPP-4 inhibition ameliorates kidney fibrosis in streptozotocin-induced diabetic mice by inhibiting endothelial-to-mesenchymal transition in a therapeutic regimen. Diabetes 63: 2120–2131, 2014. [DOI] [PubMed] [Google Scholar]
  • 60.Kanwar YS, Sun L, Xie P, Liu FY, Chen S. A glimpse of various pathogenetic mechanisms of diabetic nephropathy. Annu Rev Pathol 6: 395–423, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kato M, Arce L, Wang M, Putta S, Lanting L, Natarajan R. A microRNA circuit mediates transforming growth factor-beta1 autoregulation in renal glomerular mesangial cells. Kidney Int 80: 358–368, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kato M, Castro NE, Natarajan R. MicroRNAs: potential mediators and biomarkers of diabetic complications. Free Radic Biol Med 64: 85–94, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kato M, Dang V, Wang M, Park JT, Deshpande S, Kadam S, Mardiros A, Zhan Y, Oettgen P, Putta S, Yuan H, Lanting L, Natarajan R. TGF-beta induces acetylation of chromatin and of Ets-1 to alleviate repression of miR-192 in diabetic nephropathy. Sci Signal 6: ra43, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kato M, Natarajan R. Diabetic nephropathy—emerging epigenetic mechanisms. Nat Rev Nephrol 10: 517–530, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kato M, Natarajan R. MicroRNA circuits in transforming growth factor-beta actions and diabetic nephropathy. Semin Nephrol 32: 253–260, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kato M, Natarajan R. MicroRNAs in diabetic nephropathy: functions, biomarkers, and therapeutic targets. Ann NY Acad Sci 1353: 72–88, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kato M, Putta S, Wang M, Yuan H, Lanting L, Nair I, Gunn A, Nakagawa Y, Shimano H, Todorov I, Rossi JJ, Natarajan R. TGF-beta activates Akt kinase through a microRNA-dependent amplifying circuit targeting PTEN. Nat Cell Biol 11: 881–889, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kato M, Wang L, Putta S, Wang M, Yuan H, Sun G, Lanting L, Todorov I, Rossi JJ, Natarajan R. Post-transcriptional up-regulation of Tsc-22 by Ybx1, a target of miR-216a, mediates TGF-beta-induced collagen expression in kidney cells. J Biol Chem 285: 34004–34015, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kato M, Yuan H, Xu ZG, Lanting L, Li SL, Wang M, Hu MC, Reddy MA, Natarajan R. Role of the Akt/FoxO3a pathway in TGF-beta1-mediated mesangial cell dysfunction: a novel mechanism related to diabetic kidney disease. J Am Soc Nephrol 17: 3325–3335, 2006. [DOI] [PubMed] [Google Scholar]
  • 70.Kato M, Zhang J, Wang M, Lanting L, Yuan H, Rossi JJ, Natarajan R. MicroRNA-192 in diabetic kidney glomeruli and its function in TGF-beta-induced collagen expression via inhibition of E-box repressors. Proc Natl Acad Sci USA 104: 3432–3437, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Koga K, Yokoi H, Mori K, Kasahara M, Kuwabara T, Imamaki H, Ishii A, Mori KP, Kato Y, Ohno S, Toda N, Saleem MA, Sugawara A, Nakao K, Yanagita M, Mukoyama M. MicroRNA-26a inhibits TGF-beta-induced extracellular matrix protein expression in podocytes by targeting CTGF and is downregulated in diabetic nephropathy. Diabetologia 58: 2169–2180, 2015. [DOI] [PubMed] [Google Scholar]
  • 72.Kriegel AJ, Liu Y, Cohen B, Usa K, Liu Y, Liang M. MiR-382 targeting of kallikrein 5 contributes to renal inner medullary interstitial fibrosis. Physiol Genomics 44: 259–267, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kriegel AJ, Liu Y, Fang Y, Ding X, Liang M. The miR-29 family: genomics, cell biology, and relevance to renal and cardiovascular injury. Physiol Genomics 44: 237–244, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Krupa A, Jenkins R, Luo DD, Lewis A, Phillips A, Fraser D. Loss of microRNA-192 promotes fibrogenesis in diabetic nephropathy. J Am Soc Nephrol 21: 438–447, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, Stoffel M. Silencing of microRNAs in vivo with ‘antagomirs’. Nature 438: 685–689, 2005. [DOI] [PubMed] [Google Scholar]
  • 76.Kumar S, Liu J, McMahon AP. Defining the acute kidney injury and repair transcriptome. Semin Nephrol 34: 404–417, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Lai JY, Luo J, O'Connor C, Jing X, Nair V, Ju W, Randolph A, Ben-Dov IZ, Matar RN, Briskin D, Zavadil J, Nelson RG, Tuschl T, Brosius FC 3rd, Kretzler M, Bitzer M. MicroRNA-21 in glomerular injury. J Am Soc Nephrol 26: 805–816, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Lan YF, Chen HH, Lai PF, Cheng CF, Huang YT, Lee YC, Chen TW, Lin H. MicroRNA-494 reduces ATF3 expression and promotes AKI. J Am Soc Nephrol 23: 2012–2023, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lee SO, Masyuk T, Splinter P, Banales JM, Masyuk A, Stroope A, Larusso N. MicroRNA15a modulates expression of the cell-cycle regulator Cdc25A and affects hepatic cystogenesis in a rat model of polycystic kidney disease. J Clin Invest 118: 3714–3724, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Li Z, Rana TM. Therapeutic targeting of microRNAs: current status and future challenges. Nat Rev Drug Discov 13: 622–638, 2014. [DOI] [PubMed] [Google Scholar]
  • 81.Lin S, Gregory RI. MicroRNA biogenesis pathways in cancer. Nat Rev Cancer 15: 321–333, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lindow M, Kauppinen S. Discovering the first microRNA-targeted drug. J Cell Biol 199: 407–412, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Linkermann A, Chen G, Dong G, Kunzendorf U, Krautwald S, Dong Z. Regulated cell death in AKI. J Am Soc Nephrol 25: 2689–2701, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Liu X, Dong C, Jiang Z, Wu WK, Chan MT, Zhang J, Li H, Qin K, Sun X. MicroRNA-10b downregulation mediates acute rejection of renal allografts by derepressing BCL2L11. Exp Cell Res 333: 155–163, 2015. [DOI] [PubMed] [Google Scholar]
  • 85.Liu Y, Taylor NE, Lu L, Usa K, Cowley AW Jr, Ferreri NR, Yeo NC, Liang M. Renal medullary microRNAs in Dahl salt-sensitive rats: miR-29b regulates several collagens and related genes. Hypertension 55: 974–982, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Long J, Badal SS, Wang Y, Chang BH, Rodriguez A, Danesh FR. MicroRNA-22 is a master regulator of bone morphogenetic protein-7/6 homeostasis in the kidney. J Biol Chem 288: 36202–36214, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Long J, Wang Y, Wang W, Chang BH, Danesh FR. Identification of microRNA-93 as a novel regulator of vascular endothelial growth factor in hyperglycemic conditions. J Biol Chem 285: 23457–23465, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Long J, Wang Y, Wang W, Chang BH, Danesh FR. MicroRNA-29c is a signature microRNA under high glucose conditions that targets Sprouty homolog 1, and its in vivo knockdown prevents progression of diabetic nephropathy. J Biol Chem 286: 11837–11848, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lorenzen JM, Kaucsar T, Schauerte C, Schmitt R, Rong S, Hubner A, Scherf K, Fiedler J, Martino F, Kumarswamy R, Kolling M, Sorensen I, Hinz H, Heineke J, van Rooij E, Haller H, Thum T. MicroRNA-24 antagonism prevents renal ischemia reperfusion injury. J Am Soc Nephrol 25: 2717–2729, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lorenzen JM, Kielstein JT, Hafer C, Gupta SK, Kumpers P, Faulhaber-Walter R, Haller H, Fliser D, Thum T. Circulating miR-210 predicts survival in critically ill patients with acute kidney injury. Clin J Am Soc Nephrol 6: 1540–1546, 2011. [DOI] [PubMed] [Google Scholar]
  • 91.Lu J, Kwan BC, Lai FM, Tam LS, Li EK, Chow KM, Wang G, Li PK, Szeto CC. Glomerular and tubulointerstitial miR-638, miR-198 and miR-146a expression in lupus nephritis. Nephrology (Carlton) 17: 346–351, 2012. [DOI] [PubMed] [Google Scholar]
  • 92.Luk CC, Chow KM, Kwok JS, Kwan BC, Chan MH, Lai KB, Lai FM, Wang G, Li PK, Szeto CC. Urinary biomarkers for the prediction of reversibility in acute-on-chronic renal failure. Dis Markers 34: 179–185, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Lv C, Zhou Yh Wu C, Shao Y, Lu Cl, Wang Qy. The changes in miR-130b levels in human serum and the correlation with the severity of diabetic nephropathy. Diabetes Metab Res Rev 31: 717–724, 2015. [DOI] [PubMed] [Google Scholar]
  • 94.Mahimainathan L, Das F, Venkatesan B, Choudhury GG. Mesangial cell hypertrophy by high glucose is mediated by downregulation of the tumor suppressor PTEN. Diabetes 55: 2115–2125, 2006. [DOI] [PubMed] [Google Scholar]
  • 95.Maluf DG, Dumur CI, Suh JL, Scian MJ, King AL, Cathro H, Lee JK, Gehrau RC, Brayman KL, Gallon L, Mas VR. The urine microRNA profile may help monitor post-transplant renal graft function. Kidney Int 85: 439–449, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Martino F, Lorenzen J, Schmidt J, Schmidt M, Broll M, Gorzig Y, Kielstein JT, Thum T. Circulating microRNAs are not eliminated by hemodialysis. PLoS One 7: e38269, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Mayr C, Hemann MT, Bartel DP. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 315: 1576–1579, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Mendell JT, Olson EN. MicroRNAs in stress signaling and human disease. Cell 148: 1172–1187, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Mestecky J, Raska M, Julian BA, Gharavi AG, Renfrow MB, Moldoveanu Z, Novak L, Matousovic K, Novak J. IgA nephropathy: molecular mechanisms of the disease. Annu Rev Pathol 8: 217–240, 2013. [DOI] [PubMed] [Google Scholar]
  • 100.Meyer TW, Bennett PH, Nelson RG. Podocyte number predicts long-term urinary albumin excretion in Pima Indians with type II diabetes and microalbuminuria. Diabetologia 42: 1341–1344, 1999. [DOI] [PubMed] [Google Scholar]
  • 101.Mu J, Pang Q, Guo YH, Chen JG, Zeng W, Huang YJ, Zhang J, Feng B. Functional Implications of microRNA-215 in TGF-beta1-induced phenotypic transition of mesangial cells by targeting CTNNBIP1. PLoS One 8: e58622, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Muratsu-Ikeda S, Nangaku M, Ikeda Y, Tanaka T, Wada T, Inagi R. 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]
  • 103.Naraba H, Iwai N. Assessment of the microRNA system in salt-sensitive hypertension. Hypertens Res 28: 819–826, 2005. [DOI] [PubMed] [Google Scholar]
  • 104.Nassirpour R, Mathur S, Gosink MM, Li Y, Shoieb AM, Wood J, O'Neil SP, Homer BL, Whiteley LO. Identification of tubular injury microRNA biomarkers in urine: comparison of next-generation sequencing and qPCR-based profiling platforms. BMC Genom 15: 485, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Nathan DM, Bayless M, Cleary P, Genuth S, Gubitosi-Klug R, Lachin JM, Lorenzi G, Zinman B, Group DER. Diabetes control and complications trial/epidemiology of diabetes interventions and complications study at 30 years: advances and contributions. Diabetes 62: 3976–3986, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Neal CS, Michael MZ, Pimlott LK, Yong TY, Li JY, Gleadle JM. Circulating microRNA expression is reduced in chronic kidney disease. Nephrol Dial Transplant 26: 3794–3802, 2011. [DOI] [PubMed] [Google Scholar]
  • 107.Pagtalunan ME, Miller PL, Jumping-Eagle S, Nelson RG, Myers BD, Rennke HG, Coplon NS, Sun L, Meyer TW. Podocyte loss and progressive glomerular injury in type II diabetes. J Clin Invest 99: 342–348, 1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Pandey P, Qin S, Ho J, Zhou J, Kreidberg JA. Systems biology approach to identify transcriptome reprogramming and candidate microRNA targets during the progression of polycystic kidney disease. BMC Syst Biol 5: 56, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Park JT, Kato M, Lanting L, Castro N, Nam BY, Wang M, Kang SW, Natarajan R. Repression of let-7 by transforming growth factor-β1-induced Lin28 upregulates collagen expression in glomerular mesangial cells under diabetic conditions. Am J Physiol Renal Physiol 307: F1390–F1403, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Park JT, Kato M, Yuan H, Castro N, Lanting L, Wang M, Natarajan R. FOG2 protein down-regulation by transforming growth factor-beta1-induced microRNA-200b/c leads to Akt kinase activation and glomerular mesangial hypertrophy related to diabetic nephropathy. J Biol Chem 288: 22469–22480, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Patel A, MacMahon S, Chalmers J, Neal B, Billot L, Woodward M, Marre M, Cooper M, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Liu L, Mancia G, Mogensen CE, Pan C, Poulter N, Rodgers A, Williams B, Bompoint S, de Galan BE, Joshi R, Travert F, Group AC. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 358: 2560–2572, 2008. [DOI] [PubMed] [Google Scholar]
  • 112.Patel V, Williams D, Hajarnis S, Hunter R, Pontoglio M, Somlo S, Igarashi P. miR-17∼92 miRNA cluster promotes kidney cyst growth in polycystic kidney disease. Proc Natl Acad Sci USA 110: 10765–10770, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.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]
  • 114.Peng R, Liu H, Peng H, Zhou J, Zha H, Chen X, Zhang L, Sun Y, Yin P, Wen L, Wu T, Zhang Z. Promoter hypermethylation of let-7a-3 is relevant to its down-expression in diabetic nephropathy by targeting UHRF1. Gene 570: 57–63, 2015. [DOI] [PubMed] [Google Scholar]
  • 115.Pezzolesi MG, Satake E, McDonnell KP, Major M, Smiles AM, Krolewski AS. Circulating TGF-beta1-regulated miRNAs and the risk of rapid progression to ESRD in type 1 diabetes. Diabetes 64: 3285–3293, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Phua YL, Ho J. MicroRNAs in the pathogenesis of cystic kidney disease. Curr Opin Pediatr 27: 219–226, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Piazzon N, Maisonneuve C, Guilleret I, Rotman S, Constam DB. Bicc1 links the regulation of cAMP signaling in polycystic kidneys to microRNA-induced gene silencing. J Mol Cell Biol 4: 398–408, 2012. [DOI] [PubMed] [Google Scholar]
  • 118.Poncelet AC, Schnaper HW. Sp1 and Smad proteins cooperate to mediate transforming growth factor-beta 1-induced alpha 2(I) collagen expression in human glomerular mesangial cells. J Biol Chem 276: 6983–6992, 2001. [DOI] [PubMed] [Google Scholar]
  • 119.Putta S, Lanting L, Sun G, Lawson G, Kato M, Natarajan R. Inhibiting microRNA-192 ameliorates renal fibrosis in diabetic nephropathy. J Am Soc Nephrol 23: 458–469, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Qian Y, Feldman E, Pennathur S, Kretzler M, Brosius FC 3rd. From fibrosis to sclerosis: mechanisms of glomerulosclerosis in diabetic nephropathy. Diabetes 57: 1439–1445, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Ranganathan P, Jayakumar C, Tang Y, Park KM, Teoh JP, Su H, Li J, Kim IM, Ramesh G. 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]
  • 122.Roberts AB, McCune BK, Sporn MB. TGF-beta: regulation of extracellular matrix. Kidney Int 41: 557–559, 1992. [DOI] [PubMed] [Google Scholar]
  • 123.Ruggenenti P, Cravedi P, Remuzzi G. The RAAS in the pathogenesis and treatment of diabetic nephropathy. Nat Rev Nephrol 6: 319–330, 2010. [DOI] [PubMed] [Google Scholar]
  • 124.Saikumar J, Hoffmann D, Kim TM, Gonzalez VR, Zhang Q, Goering PL, Brown RP, Bijol V, Park PJ, Waikar SS, Vaidya VS. 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]
  • 125.Sampson VB, Rong NH, Han J, Yang Q, Aris V, Soteropoulos P, Petrelli NJ, Dunn SP, Krueger LJ. MicroRNA let-7a down-regulates MYC and reverts MYC-induced growth in Burkitt lymphoma cells. Cancer Res 67: 9762–9770, 2007. [DOI] [PubMed] [Google Scholar]
  • 126.Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, Culleton B, Hamm LL, McCullough PA, Kasiske BL, Kelepouris E, Klag MJ, Parfrey P, Pfeffer M, Raij L, Spinosa DJ, Wilson PW. Kidney disease as a risk factor for development of cardiovascular disease: a statement from the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation 108: 2154–2169, 2003. [DOI] [PubMed] [Google Scholar]
  • 127.Sedeek M, Montezano AC, Hebert RL, Gray SP, Di Marco E, Jha JC, Cooper ME, Jandeleit-Dahm K, Schiffrin EL, Wilkinson-Berka JL, Touyz RM. Oxidative stress, Nox isoforms and complications of diabetes—potential targets for novel therapies. J Cardiovasc Transl Res 5: 509–518, 2012. [DOI] [PubMed] [Google Scholar]
  • 128.Serino G, Sallustio F, Cox SN, Pesce F, Schena FP. Abnormal miR-148b expression promotes aberrant glycosylation of IgA1 in IgA nephropathy. J Am Soc Nephrol 23: 814–824, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Sharma K, Ziyadeh FN. Hyperglycemia and diabetic kidney disease. The case for transforming growth factor-beta as a key mediator. Diabetes 44: 1139–1146, 1995. [DOI] [PubMed] [Google Scholar]
  • 130.Shi S, Yu L, Chiu C, Sun Y, Chen J, Khitrov G, Merkenschlager M, Holzman LB, Zhang W, Mundel P, Bottinger EP. Podocyte-selective deletion of dicer induces proteinuria and glomerulosclerosis. J Am Soc Nephrol 19: 2159–2169, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Sole C, Cortes-Hernandez J, Felip ML, Vidal M, Ordi-Ros J. miR-29c in urinary exosomes as predictor of early renal fibrosis in lupus nephritis. Nephrol Dial Transplant 30: 14898–1496, 2015. [DOI] [PubMed] [Google Scholar]
  • 132.Sui W, Liu F, Chen J, Ou M, Dai Y. Microarray technology for analysis of microRNA expression in renal biopsies of lupus nephritis patients. Methods Mol Biol 1134: 211–220, 2014. [DOI] [PubMed] [Google Scholar]
  • 133.Sun H, Li QW, Lv XY, Ai JZ, Yang QT, Duan JJ, Bian GH, Xiao Y, Wang YD, Zhang Z, Liu YH, Tan RZ, Yang Y, Wei YQ, Zhou Q. MicroRNA-17 post-transcriptionally regulates polycystic kidney disease-2 gene and promotes cell proliferation. Mol Biol Rep 37: 2951–2958, 2010. [DOI] [PubMed] [Google Scholar]
  • 134.Sun L, Zhang D, Liu F, Xiang X, Ling G, Xiao L, Liu Y, Zhu X, Zhan M, Yang Y, Kondeti VK, Kanwar YS. Low-dose paclitaxel ameliorates fibrosis in the remnant kidney model by down-regulating miR-192. J Pathol 225: 364–377, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Sun Y, Koo S, White N, Peralta E, Esau C, Dean NM, Perera RJ. Development of a micro-array to detect human and mouse microRNAs and characterization of expression in human organs. Nucleic Acids Res 32: e188, 2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Szeto CC, Ching-Ha KB, Ka-Bik L, Mac-Moune LF, Cheung-Lung CP, Gang W, Kai-Ming C, Kam-Tao LP. 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]
  • 137.Szeto CC, Li PK. MicroRNAs in IgA nephropathy. Nat Rev Nephrol 10: 249–256, 2014. [DOI] [PubMed] [Google Scholar]
  • 138.Tian Z, Greene AS, Pietrusz JL, Matus IR, Liang M. MicroRNA-target pairs in the rat kidney identified by microRNA microarray, proteomic, and bioinformatic analysis. Genome Res 18: 404–411, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Tijsen AJ, Creemers EE, Moerland PD, de Windt LJ, van der Wal AC, Kok WE, Pinto YM. MiR423-5p as a circulating biomarker for heart failure. Circ Res 106: 1035–1039, 2010. [DOI] [PubMed] [Google Scholar]
  • 140.Tran U, Zakin L, Schweickert A, Agrawal R, Doger R, Blum M, De Robertis EM, Wessely O. The RNA-binding protein bicaudal C regulates polycystin 2 in the kidney by antagonizing miR-17 activity. Development 137: 1107–1116, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Trionfini P, Benigni A, Remuzzi G. MicroRNAs in kidney physiology and disease. Nat Rev Nephrol 11: 23–33, 2015. [DOI] [PubMed] [Google Scholar]
  • 142.Tsuchida K, Zhu Y, Siva S, Dunn SR, Sharma K. Role of Smad4 on TGF-beta-induced extracellular matrix stimulation in mesangial cells. Kidney Int 63: 2000–2009, 2003. [DOI] [PubMed] [Google Scholar]
  • 143.van den Akker EK, Dor FJ, JNIJ, de Bruin RW. MicroRNAs in kidney transplantation: living up to their expectations? J Transplant 2015: 354826, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.van Rooij E, Kauppinen S. Development of microRNA therapeutics is coming of age. EMBO Mol Med 6: 851–864, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Wang B, Herman-Edelstein M, Koh P, Burns W, Jandeleit-Dahm K, Watson A, Saleem M, Goodall GJ, Twigg SM, Cooper ME, Kantharidis P. E-cadherin expression is regulated by miR-192/215 by a mechanism that is independent of the profibrotic effects of transforming growth factor-beta. Diabetes 59: 1794–1802, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Wang B, Jha JC, Hagiwara S, McClelland AD, Jandeleit-Dahm K, Thomas MC, Cooper ME, Kantharidis P. Transforming growth factor-beta1-mediated renal fibrosis is dependent on the regulation of transforming growth factor receptor 1 expression by let-7b. Kidney Int 85: 352–361, 2014. [DOI] [PubMed] [Google Scholar]
  • 147.Wang B, Koh P, Winbanks C, Coughlan MT, McClelland A, Watson A, Jandeleit-Dahm K, Burns WC, Thomas MC, Cooper ME, Kantharidis P. miR-200a prevents renal fibrogenesis through repression of TGF-beta2 expression. Diabetes 60: 280–287, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Wang B, Komers R, Carew R, Winbanks CE, Xu B, Herman-Edelstein M, Koh P, Thomas M, Jandeleit-Dahm K, Gregorevic P, Cooper ME, Kantharidis P. Suppression of microRNA-29 expression by TGF-beta1 promotes collagen expression and renal fibrosis. J Am Soc Nephrol 23: 252–265, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Wang E, Hsieh-Li HM, Chiou YY, Chien YL, Ho HH, Chin HJ, Wang CK, Liang SC, Jiang ST. Progressive renal distortion by multiple cysts in transgenic mice expressing artificial microRNAs against Pkd1. J Pathol 222: 238–248, 2010. [DOI] [PubMed] [Google Scholar]
  • 150.Wang G, Kwan BC, Lai FM, Choi PC, Chow KM, Li PK, Szeto CC. Intrarenal expression of miRNAs in patients with hypertensive nephrosclerosis. Am J Hypertens 23: 78–84, 2010. [DOI] [PubMed] [Google Scholar]
  • 151.Wang G, Kwan BC, Lai FM, Chow KM, Li PK, Szeto CC. Urinary miR-21, miR-29, and miR-93: novel biomarkers of fibrosis. Am J Nephrol 36: 412–418, 2012. [DOI] [PubMed] [Google Scholar]
  • 152.Wang G, Kwan BC, Lai FM, Chow KM, Li PK, Szeto CC. Urinary sediment miRNA levels in adult nephrotic syndrome. Clin Chim Acta 418: 5–11, 2013. [DOI] [PubMed] [Google Scholar]
  • 153.Wang J, Gao Y, Ma M, Li M, Zou D, Yang J, Zhu Z, Zhao X. Effect of miR-21 on renal fibrosis by regulating MMP-9 and TIMP1 in kk-ay diabetic nephropathy mice. Cell Biochem Biophys 67: 537–546, 2013. [DOI] [PubMed] [Google Scholar]
  • 154.Wang K, Zhang S, Marzolf B, Troisch P, Brightman A, Hu Z, Hood LE, Galas DJ. Circulating microRNAs, potential biomarkers for drug-induced liver injury. Proc Natl Acad Sci USA 106: 4402–4407, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Wang N, Zhou Y, Jiang L, Li D, Yang J, Zhang CY, Zen K. 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]
  • 156.Wang Q, Wang Y, Minto AW, Wang J, Shi Q, Li X, Quigg RJ. MicroRNA-377 is up-regulated and can lead to increased fibronectin production in diabetic nephropathy. FASEB J 22: 4126–4135, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Wang XX, Jiang T, Shen Y, Caldas Y, Miyazaki-Anzai S, Santamaria H, Urbanek C, Solis N, Scherzer P, Lewis L, Gonzalez FJ, Adorini L, Pruzanski M, Kopp JB, Verlander JW, Levi M. Diabetic nephropathy is accelerated by farnesoid X receptor deficiency and inhibited by farnesoid X receptor activation in a type 1 diabetes model. Diabetes 59: 2916–2927, 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Wei Q, Bhatt K, He HZ, Mi QS, Haase VH, Dong Z. 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]
  • 159.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]
  • 160.Wilflingseder J, Sunzenauer J, Toronyi E, Heinzel A, Kainz A, Mayer B, Perco P, Telkes G, Langer RM, Oberbauer R. Molecular pathogenesis of post-transplant acute kidney injury: assessment of whole-genome mRNA and miRNA profiles. PLoS One 9: e104164, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Winter J, Jung S, Keller S, Gregory RI, Diederichs S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat Cell Biol 11: 228–234, 2009. [DOI] [PubMed] [Google Scholar]
  • 162.Wu J, Zheng C, Fan Y, Zeng C, Chen Z, Qin W, Zhang C, Zhang W, Wang X, Zhu X, Zhang M, Zen K, Liu Z. Downregulation of microRNA-30 facilitates podocyte injury and is prevented by glucocorticoids. J Am Soc Nephrol 25: 92–104, 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Wyatt RJ, Julian BA. IgA nephropathy. N Engl J Med 368: 2402–2414, 2013. [DOI] [PubMed] [Google Scholar]
  • 164.Xu X, Kriegel AJ, Liu Y, Usa K, Mladinov D, Liu H, Fang Y, Ding X, Liang M. Delayed ischemic preconditioning contributes to renal protection by upregulation of miR-21. Kidney Int 82: 1167–1175, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Yamamoto T, Nakamura T, Noble NA, Ruoslahti E, Border WA. Expression of transforming growth factor beta is elevated in human and experimental diabetic nephropathy. Proc Natl Acad Sci USA 90: 1814–1818, 1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Yang Y, Xiao L, Li J, Kanwar YS, Liu F, Sun L. Urine miRNAs: potential biomarkers for monitoring progression of early stages of diabetic nephropathy. Med Hypotheses 81: 274–278, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Zhang C, Zeng X, Li Z, Wang Z, Li S. Immunoglobulin A nephropathy: current progress and future directions. Transl Res 166: 134–144, 2015. [DOI] [PubMed] [Google Scholar]
  • 168.Zhang Y, Feng X, We R, Derynck R. Receptor-associated Mad homologues synergize as effectors of the TGF-beta response. Nature 383: 168–172, 1996. [DOI] [PubMed] [Google Scholar]
  • 169.Zhang Z, Peng H, Chen J, Chen X, Han F, Xu X, He X, Yan N. MicroRNA-21 protects from mesangial cell proliferation induced by diabetic nephropathy in db/db mice. FEBS Lett 583: 2009–2014, 2009. [DOI] [PubMed] [Google Scholar]
  • 170.Zhong X, Chung ACK, Chen HY, Dong Y, Meng XM, Li R, Yang W, Hou FF, Lan HY. miR-21 is a key therapeutic target for renal injury in a mouse model of type 2 diabetes. Diabetologia 56: 663–674, 2013. [DOI] [PubMed] [Google Scholar]
  • 171.Zhu Y, Usui HK, Sharma K. Regulation of transforming growth factor beta in diabetic nephropathy: implications for treatment. Semin Nephrol 27: 153–160, 2007. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Renal Physiology are provided here courtesy of American Physiological Society

RESOURCES