Elsevier

Cellular Signalling

Volume 71, July 2020, 109548
Cellular Signalling

Modulation of polycystic kidney disease by non-coding RNAs

https://doi.org/10.1016/j.cellsig.2020.109548Get rights and content

Abstract

Purpose of review

microRNAs (miRNAs) are a class of small, evolutionarily conserved, non-coding RNAs (ncRNAs) that function as inhibitors of post-transcriptional mRNA expression. They are implicated in the pathogenesis of numerous diseases, including many common kidney conditions. In this review, we focus on how miRNAs impact autosomal dominant polycystic kidney disease (ADPKD) progression. We also discuss the feasibility of the emerging novel antisense oligonucleotides (ASOs) drug class, which includes anti-miRNA drugs, for the treatment of ADPKD.

Recent findings

Aberrant miRNA expression is observed in multiple PKD murine models and human ADPKD samples. Gain and loss-of-function studies have directly linked dysregulated miRNA activity to kidney cyst growth. The most comprehensively studied miRNA in PKD is the miR-17 family, which promotes PKD progression through the rewiring of cyst metabolism and by directly inhibiting PKD1 and PKD2 expression. This discovery has led to the development of an anti-miR-17 drug for ADPKD treatment. Other miRNAs such as miR-21, miR-193, and miR-214 are also known to regulate cyst growth by modulating cyst epithelial apoptosis, proliferation, and interstitial inflammation.

Summary

miRNAs have emerged as novel pathogenic regulators of ADPKD progression. Anti-miR-based drugs represent a new therapeutic modality to treat ADPKD patients.

Introduction

The vast majority of the genome contains regions that do not code for proteins [1]. Historically termed as the ‘junk DNA,’ it is now known that a significant portion of this DNA undergoes transcription to give rise to non-coding RNAs (ncRNAs) with pivotal functions [1]. These RNAs, as the name suggests, do not encode for proteins but rather remain as RNAs during their lifetime and regulate gene/mRNA expression and mRNA metabolism.

ncRNAs are of two types - long ncRNAs (lncRNAs) that are >200 nucleotides in length and short ncRNAs of <30 nucleotides in length [2]. There are three major classes of short ncRNAs - miRNAs, siRNAs, and piRNAs [3]. Amongst the ncRNAs, miRNAs the most are widely studied. Compelling evidence supports the idea that miRNAs are novel regulators of disease pathogenesis [4,5]. First, aberrant miRNA expression/function is observed in numerous diseases such as cancer, tissue fibrosis, congenital and developmental conditions, and various monogenetic disorders including polycystic kidney disease (PKD). Second, gain and loss-of-function approaches causally link abnormal activity of many miRNAs to the progression of these diseases. Third, the pathogenic miRNAs contribute to the dysregulation of large gene networks in various diseases. Finally, these observations have led to the realization that miRNAs are novel drug targets for the treatment of many complex diseases. Indeed, multiple miRNA-based drugs have now advanced to human clinical trials for the treatment of cancer, tissue fibrosis, and two monogenic kidney diseases – Alport syndrome and autosomal dominant polycystic kidney disease (ADPKD).

PKD is amongst the most common human monogenic disease caused primarily by mutations in PKD1 or PKD2. The clinical hallmark of this disorder is the massive bilateral kidney enlargement due to innumerable cysts. The cysts are lined by hyperproliferative and hypersecretory cyst epithelial cells, which cause uncontrolled cyst growth and expansion, ultimately resulting in kidney failure. Several signaling pathways, including cAMP, mTOR, and cMyc have been known for a long time to drive PKD progression [6]. Recent work indicates that miRNA-mediated signaling is a key new driver of PKD pathogenesis [[7], [8], [9], [10]]. This review focuses on the progress made so far in our understanding of the role of miRNAs in PKD with an emphasis on the therapeutic targeting of these ncRNAs in PKD treatment.

Section snippets

miRNA biogenesis and mechanism of action

miRNAs are ~22 nucleotides double-stranded RNA molecules and were first discovered in 1993 in C.elegans as regulators of larvae patterning [11,12]. Since this initial discovery, it has become clear that miRNAs are integral to virtually all aspects of mammalian biology ranging from the initial stages of embryo formation, stem cell biology and organ development to immune response, tissue injury/repair, aging, and disease progression [13,14].

miRNAs are produced either from intragenic (located

miRNAs in kidney development and homeostasis

The embryonic kidney consists of two precursor tissues: the metanephric mesenchyme and the ureteric bud [[21], [22], [23]]. Molecular signals from the ureteric bud induce the nephron progenitor cells of the metanephric mesenchyme to undergo differentiation into renal vesicles, which eventually give rise to glomeruli and nephrons [21,22]. Conversely, the metanephric mesenchyme induces branching morphogenesis of the ureteric bud, which differentiates into the collecting duct network [23]. During

miRNAs: new regulators of PKD pathogenesis

Numerous miRNAs are aberrantly expressed in murine and human forms of ADPKD. However, only a few have been causally linked to ADPKD progression. In the following sections, we discuss two cyst-promoting (miR-21 and miR-17) and cyst-suppressing (miR-193 and miR-214) miRNAs.

miR-21

miR-21 is an evolutionarily conserved and an extensively studied oncogenic miRNA [41]. The miR-21 gene is located within the protein-coding TMEM49 gene; however, it is transcribed independently via its own promoter [42] (Fig. 2). Multiple genomic regions in TMEM49 introns function as putative promoter regions of miR-21 [43,44]. miR-21 is broadly expressed in many cell types during development and, this includes the kidney epithelium and podocytes. Surprisingly, despite the widespread expression

miR-17–92 cluster

The miR-17–92 cluster was amongst the first miRNAs found to be amplified in cancerous tissues earning it the moniker ‘oncomir-1’ [65]. This finding sparked an intense interest in understanding its role in development and disease. miR-17–92 is a polycistronic miRNA cluster, which produces six individual miRNAs (miR-17, miR-18a, miR-19a, miR-20a, miR-19b-1, and miR-92a) [66,67]. The cluster is located within a 7 kb non-protein coding gene MIR17HG or C13orf25 (Fig. 2) and is evolutionarily

Cyst suppressing miRNAs

miR193-3p is a tumor suppressor miRNA [[83], [84], [85], [86]]. During ADPKD progression, its activity is suppressed [87]. Parallel microarray analysis for miRNA and mRNA expression on cell lines derived from normal kidneys and human ADPKD kidneys has shown that miR193b-3p is one of two significantly downregulated miRNAs. This miRNA functions by inhibiting several factors that regulate cell proliferation, such as Erb-B2 Receptor Tyrosine Kinase 4 (ErbB4) [[88], [89], [90], [91]]. ErbB4 activity

Pharmacological modulation of miRNA activity

RNA-based drugs predate the discovery of miRNAs by a couple of decades. The RNA-based approach is applied in two ways: first, is the antisense oligonucleotide (ASO)-based approach [[93], [94], [95], [96]]. ASOs bind to target mRNAs and physically prevent ribosomes from translating the mRNA. In this way, the ASOs act as steric inhibitors of mRNA translation. ASOs can also be designed such that upon binding to target mRNAs, they recruit RNAase H, which degrades the mRNA [97]. The second approach

miRNA-based therapeutics for PKD

The first miRNA drug to be tested in humans was Mirvirasen, an LNA-modified anti-miR-122, for the treatment of Hepatitis C (HCV) [110]. In a phase 2A clinical trial, this drug demonstrated remarkable efficacy in suppressing HCV [111]. Although a follow-up clinical trial has not yet been performed, Miravirasen has provided the initial clinical proof of concept efficacy for the miRNA-based therapeutics platform. Several other miRNA-based drugs have now entered human clinical development for

Conclusion

ADPKD is the most common genetic cause of renal failure. Current treatment options are limited. miRNAs have emerged as promising drug targets in ADPKD. The biological effects of three epithelial miRNAs (miR-17–92, miR-21 and miR-193b-3p) and one stromal miRNA (miR-214) have been evaluated in ADPKD. miR-17–92 and miR-21 promote disease progression in ADPKD. Conversely, miR-193b-3p, and miR-214 attenuate cyst growth. These studies have led to the development of miRNA-based drugs for the treatment

Credit authors

Harini Ramalingam, Matanel Yheskel, and Vishal Patel wrote this review.

Declaration of Competing Interest

Vishal Patel has applied for a patent related to the treatment of polycystic kidney disease using miR-17 inhibitors. The Patel lab has a sponsored research agreement with Regulus Therapeutics. The remaining authors declare no conflict of interests.

Acknowledgements

The work from the authors laboratory is supported by National Institute of Health (R01DK102572) and the Department of Defense (D01 W81XWH1810673) to Vishal Patel. Harini Ramalingam is supported by the PKD Foundation Fellowship Grant.

References (114)

  • S. Fujita

    miR-21 gene expression triggered by AP-1 is sustained through a double-negative feedback mechanism

    J. Mol. Biol.

    (2008)
  • D. Loffler

    Interleukin-6 dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer

    Blood

    (2007)
  • M.E. Hatley

    Modulation of K-Ras-dependent lung tumorigenesis by MicroRNA-21

    Cancer Cell

    (2010)
  • J.T. Mendell

    miRiad roles for the miR-17-92 cluster in development and disease

    Cell

    (2008)
  • A. Tanzer et al.

    Molecular evolution of a microRNA cluster

    J. Mol. Biol.

    (2004)
  • A. Ventura

    Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters

    Cell

    (2008)
  • J. Wang

    Bmp signaling regulates myocardial differentiation from cardiac progenitors through a MicroRNA-mediated mechanism

    Dev. Cell

    (2010)
  • Y. Lu et al.

    Transgenic over-expression of the microRNA miR-17-92 cluster promotes proliferation and inhibits differentiation of lung epithelial progenitor cells

    Dev. Biol.

    (2007)
  • F. Zeng et al.

    Deletion of ErbB4 accelerates polycystic kidney disease progression in cpk mice

    Kidney Int.

    (2014)
  • D.A. Loebel et al.

    A conserved noncoding intronic transcript at the mouse Dnm3 locus

    Genomics

    (2005)
  • Y. Dong et al.

    Strategies, design, and chemistry in siRNA delivery systems

    Adv. Drug Deliv. Rev.

    (2019)
  • S.T. Crooke et al.

    RNA-targeted therapeutics

    Cell Metab.

    (2018)
  • M.C. Frith et al.

    The amazing complexity of the human transcriptome

    Eur. J. Hum. Genet.

    (2005)
  • S. Hombach et al.

    Non-coding RNAs: classification, biology and functioning

    Adv. Exp. Med. Biol.

    (2016)
  • V.N. Kim et al.

    Biogenesis of small RNAs in animals

    Nat. Rev. Mol. Cell Biol.

    (2009)
  • P.C. Harris et al.

    Genetic mechanisms and signaling pathways in autosomal dominant polycystic kidney disease

    J. Clin. Invest.

    (2014)
  • V. Patel

    miR-17~92 miRNA cluster promotes kidney cyst growth in polycystic kidney disease

    Proc. Natl. Acad. Sci. U. S. A.

    (2013)
  • S. Hajarnis

    microRNA-17 family promotes polycystic kidney disease progression through modulation of mitochondrial metabolism

    Nat. Commun.

    (2017)
  • R. Lakhia

    MicroRNA-21 aggravates cyst growth in a model of polycystic kidney disease

    J. Am. Soc. Nephrol.

    (2016)
  • R. Lakhia

    PPARalpha agonist fenofibrate enhances fatty acid beta-oxidation and attenuates polycystic kidney and liver disease in mice

    Am. J. Physiol. Ren. Physiol.

    (2018)
  • I. Alvarez-Garcia et al.

    MicroRNA functions in animal development and human disease

    Development

    (2005)
  • D. Sayed et al.

    MicroRNAs in development and disease

    Physiol. Rev.

    (2011)
  • A. Rodriguez et al.

    Identification of mammalian microRNA host genes and transcription units

    Genome Res.

    (2004)
  • L.F. Gulyaeva et al.

    Regulatory mechanisms of microRNA expression

    J. Transl. Med.

    (2016)
  • L. Cantini

    Identification of microRNA clusters cooperatively acting on epithelial to mesenchymal transition in triple negative breast cancer

    Nucleic Acids Res.

    (2019)
  • E.C. Lee

    Discovery and preclinical evaluation of anti-miR-17 oligonucleotide RGLS4326 for the treatment of polycystic kidney disease

    Nat. Commun.

    (2019)
  • J. O’Brien et al.

    Overview of MicroRNA biogenesis, mechanisms of actions, and circulation

    Front. Endocrinol. (Lausanne)

    (2018)
  • M. Yoda

    ATP-dependent human RISC assembly pathways

    Nat. Struct. Mol. Biol.

    (2010)
  • M.H. Little et al.

    Mammalian kidney development: principles, progress, and projections

    Cold Spring Harb. Perspect. Biol.

    (2012)
  • R.M. Kao et al.

    Invasion of distal nephron precursors associates with tubular interconnection during nephrogenesis

    J. Am. Soc. Nephrol.

    (2012)
  • W.F. Neiss

    Morphogenesis and histogenesis of the connecting tubule in the rat kidney

    Anat. Embryol. (Berl)

    (1982)
  • J.F. Bertram et al.

    Human nephron number: implications for health and disease

    Pediatr. Nephrol.

    (2011)
  • J. Ho et al.

    MicroRNAs in renal development

    Pediatr. Nephrol.

    (2013)
  • V.K. Nagalakshmi et al.

    The ureteric bud epithelium: morphogenesis and roles in metanephric kidney patterning

    Mol. Reprod. Dev.

    (2015)
  • J.Y. Chu

    Dicer function is required in the metanephric mesenchyme for early kidney development

    Am. J. Physiol. Ren. Physiol.

    (2014)
  • J. Ho

    The pro-apoptotic protein Bim is a microRNA target in kidney progenitors

    J. Am. Soc. Nephrol.

    (2011)
  • V. Patel

    MicroRNAs regulate renal tubule maturation through modulation of Pkd1

    J. Am. Soc. Nephrol.

    (2012)
  • S. Shi

    Podocyte-selective deletion of dicer induces proteinuria and glomerulosclerosis

    J. Am. Soc. Nephrol.

    (2008)
  • J. Ho

    Podocyte-specific loss of functional microRNAs leads to rapid glomerular and tubular injury

    J. Am. Soc. Nephrol.

    (2008)
  • S.J. Harvey

    Podocyte-specific deletion of dicer alters cytoskeletal dynamics and causes glomerular disease

    J. Am. Soc. Nephrol.

    (2008)
  • Cited by (21)

    • Autophagy-mediated reduction of miR-345 contributes to hepatic cystogenesis in polycystic liver disease

      2021, JHEP Reports
      Citation Excerpt :

      Direct pharmacological modulation of miRNA expression in PLD/PKD remains an undeveloped area. The anti-miR-17A antisense oligonucleotide drug, RGLS4326, decreases the level of miR-17A overexpressed in kidney, subsequently inhibiting renal cystogenesis in an animal model of PKD.39,40 Another drug that re-establishes the levels of downregulated miR-29 in fibrotic diseases, but not in PKD, is remlarsen.41,42

    • A methionine-Mettl3-N<sup>6</sup>-methyladenosine axis promotes polycystic kidney disease

      2021, Cell Metabolism
      Citation Excerpt :

      To our knowledge, the role of Mettl3 in any form of kidney disease has not been studied. Autosomal dominant polycystic kidney disease (ADPKD), primarily caused by mutations of PKD1 or PKD2, is among the most common monogenetic human disorders (Patel et al., 2009; Ramalingam et al., 2020). The defining feature are the numerous renal tubule-derived cysts, which enlarge over time, causing massive bilateral kidney enlargement.

    View all citing articles on Scopus
    View full text