Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Jul 1.
Published in final edited form as: Cell Signal. 2020 Feb 14;71:109568. doi: 10.1016/j.cellsig.2020.109568

Role of Transcription Factor Hepatocyte Nuclear Factor-1β in Polycystic Kidney Disease

Annie Shao a, Siu Chiu Chan a, Peter Igarashi a,*
PMCID: PMC7183897  NIHMSID: NIHMS1564788  PMID: 32068086

Abstract

Hepatocyte nuclear factor-1β (HNF-1β) is a DNA-binding transcription factor that is essential for normal kidney development. Mutations of HNF1B in humans produce cystic kidney diseases, including renal cysts and diabetes, multicystic dysplastic kidneys, glomerulocystic kidney disease, and autosomal dominant tubulointerstitial kidney disease. Expression of HNF1B is reduced in cystic kidneys from humans with ADPKD, and HNF1B has been identified as a modifier gene in PKD. Genome-wide analysis of chromatin binding has revealed that HNF-1β directly regulates the expression of known PKD genes, such as PKHD1 and PKD2, as well as genes involved in PKD pathogenesis, including cAMP-dependent signaling, renal fibrosis, and Wnt signaling. In addition, a role of HNF-1β in regulating the expression of noncoding RNAs (microRNAs and long noncoding RNAs) has been identified. These findings indicate that HNF-1β regulates a transcriptional and post-transcriptional network that plays a central role in renal cystogenesis.

Keywords: gene transcription, kidney development, kidney cysts, chromatin immunoprecipitation, cyclic AMP, Wnt signaling, kidney fibrosis, noncoding RNA

1. Introduction

1.1. Structure and function of HNF-1β

Hepatocyte nuclear factor-1β (HNF-1β) is a DNA-binding transcription factor that regulates tissue-specific gene expression in epithelial organs including the kidney.1 In the kidney, HNF-1β is expressed in all tubular epithelial cells composing the nephrons and collecting ducts where it controls the expression of genes involved in membrane transport, cell differentiation, and metabolism.2; 3 Human HNF-1β contains 557 amino acids4 organized into an amino-terminal dimerization domain, a bipartite DNA-binding domain, and a carboxy-terminal transactivation domain 5; 6 (Figure 1). HNF-1β binds DNA as a homodimer or as a heterodimer with its paralogue, HNF-1α.7 HNF-1α is restricted to renal proximal tubules where it regulates genes involved in amino acid, phosphate, and organic acid transport, whereas HNF-1β is expressed more broadly in all nephron segments.7 The DNA-binding domains of HNF-1α and HNF-1β are highly conserved, and the proteins recognize a similar DNA sequence. However, the carboxy-terminal domains are divergent, which enables HNF-1β homodimers and HNF-1β/HNF-1α heterodimers to interact with different coactivators and corepressors.57 Dimerization of HNF-1β and HNF-1α is mediated by their amino-terminal domains and promoted by interaction with dimerization cofactor of HNF1 (DCoH).8; 9

Figure 1. Structure of HNF-1β and cyst-causing mutations.

Figure 1.

Schematic representation of HNF-1β protein structure. The N-terminal dimerization domain (D), POU-specific (POUs), POU-homeo (POUH) domains, and C-terminal transactivation domain are shown. Locations of a subset of mutations that are referenced in this review and associated with kidney cysts are depicted above.6; 25; 26; 28; 29; 39; 52; 56; 57; 63; 70; 111

The DNA-binding domain of HNF-1β is composed of a POU (Pit-1, Oct-1/2, Unc-86)-specific domain (POUS) and POU-homeodomain (POUH). The POUS and POUH domains adopt a helix-turn-helix tertiary structure that fits into the major groove of DNA10; 11 and mediates sequence-specific binding.5; 6 HNF-1β recognizes a pseudopalindromic consensus sequence, 5′-GGTTAATNATTAAC-3′, 7; 12 that is often present in the promoter region or enhancers of target genes. Once bound to DNA, HNF-1β modulates gene expression by inducing conformational changes in chromatin structure via its interaction with co-activators and corepressors. For example, interactions of the carboxy-terminal domain of HNF-1β with the histone acetylases CREB binding protein and P300/CBP-associated factor results in transcriptional activation6 Conversely, HNF-1β can function as a transcriptional repressor through interactions of the carboxy-terminal domain with histone deacetylases.6 An unusual property of HNF-1β is that it remains bound to DNA during cell division, which differs from most transcription factors that dissociate from mitotic chromosomes.13 This “bookmarking” function enables rapid restoration of HNF-1β target gene activation in daughter cells.14

1.2. Essential role in kidney development

HNF-1β is expressed in the embryonic kidney and is required at multiple steps of kidney development, including branching morphogenesis, nephrogenesis, nephron patterning, and tubulogenesis. Kidney development involves complex reciprocal interactions and signaling between two main cellular compartments: the ureteric bud (UB) and metanephric mesenchyme (MM). In mice at embryonic day 10.5 (E10.5), the UB forms as an outgrowth from the posterior Wolffian duct (WD). This process is induced by the adjacent MM, which contains nephron progenitor cells and stromal cells.15; 16 The UB invades the MM and undergoes branching morphogenesis giving rise to the renal collecting system. The tips of the branching UB induce the MM to condense and undergo mesenchymal-to-epithelial transition (MET) forming the nephrons proper. The first polarized epithelial MM-derived structures, the renal vesicles, subsequently differentiate into the comma-shaped- and S-shaped bodies, which then fuse with the collecting system forming maturing, segmented nephrons.1518 HNF-1β is expressed in the mouse WD epithelium at E9.5 and in the branching UB throughout later stages of kidney development.1 HNF-1β is absent in the MM and first appears at the renal vesicle stage.19; 20 Expression is sustained in the mature nephron.1; 19

To identify the role of HNF-1β in kidney development, Lokmane et al. generated HNF-1β-deficient mouse embryos using tetraploid embryo complementation.19 The initial branching of the UB is delayed in mutant embryos, and branching remains defective throughout subsequent organogenesis. As a consequence, HNF-1β mutant kidneys are hypoplastic.19 One major signaling pathway that regulates branching morphogenesis is Ret/Glial cell line-derived neurotrophic factor (GDNF) signaling. The MM expresses the ligand GDNF, which engages the Ret receptor and Gfrcd co-receptor expressed in the WD and UB.15; 16 In HNF-1β mutant kidneys, the expression of both Ret and GDNF is significantly reduced.19 To further elucidate the role of Ret/GDNF signaling, Desgrange et al. ablated HNF-1β specifically in the UB of embryonic mouse kidneys using Cre-loxP recombination.21 Mutant kidneys show severely impaired branching morphogenesis21 associated with downregulation of Gfrα, indicating that HNF-1β directly regulates Ret/GDNF signaling. In addition, the expression of Pax-2, a transcription factor that regulates branching morphogenesis by activating Wnt11 expression,22 is downregulated.21 Wnt11 plays an essential role in branching morphogenesis by maintaining GDNF expression via an autoregulatory loop with Ret/GDNF.23 These findings demonstrate that HNF-1β is essential for kidney branching morphogenesis through regulation of Pax-2 and a Ret/GDNF/Wnt11 autoregulatory loop.

HNF-1β is also necessary for proper nephron induction and patterning. HNF-1β mutant kidneys show decreased expression of Wnt9b,19 the primary inducer of nephrogenesis24 and a direct downstream target of HNF-1β.19 Wnt9b is secreted by the UB tips and acts as a paracrine signal initiating nephrogenesis in the MM. In addition, HNF-1β regulates MM-autonomous signaling pathways that drive nephrogenesis. Massa et al. ablated HNF-1β in the MM of the mouse kidney using Cre-lox recombination and found that renal vesicles still form, but the expression of nephron markers is delayed and S-shaped bodies exhibit structural defects.20 The mid- and upper- limbs, which give rise to the proximal and distal convoluted tubules, are deformed and show increased apoptosis and decreased cell proliferation. At birth, the mutant kidneys contain greatly reduced proximal and distal convoluted tubules.20 These results indicate that expression of HNF-1β in the MM is not required for initial nephrogenesis but is required for the differentiation of the proximal and distal tubules.

2. Human cystic diseases associated with mutations of HNF-1β

Heterozygous mutations of HNF1B are among the most common single-gene mutations found in developmental kidney disease.25 In this section, we discuss the congenital kidney abnormalities that are associated with mutations of HNF1B focusing on cystic phenotypes.

2.1. HNF-1β mutations in human disease

More than 100 pathogenic mutations of HNF1B have been reported in humans.26 The mutations range from intragenic mutations, including missense, frameshift, nonsense, and small insertions or deletions, to whole-gene deletions and large genomic rearrangements 25; 27; 28 (Figure 1). Mutations of HNF1B are usually present in the germline but can also occur de novo28; 29 Few genotype-phenotype correlations have been identified, in part because the severity of renal disease is highly variable even within families carrying the identical mutation.28; 30 This variable expressivity is reminiscent of ADPKD and is likely due to additional genetic, environmental, and epigenetic factors modulating the HNF1B mutant phenotype.2; 29 Developmental timing may also play a role as children with very early onset disease have more severe kidney abnormalities and accelerated loss of kidney function than those with later onset.31

One-third of the reported human HNF1B mutations consist of whole-gene deletions.26 Such deletions are difficult to detect by conventional DNA sequencing, and specialized methods to measure copy number variation (CNV) must be employed. Most individuals with HNF1B deletions are heterozygous for the 17q12 deletion, which is a recurrent 1.4 Mb deletion that encompasses HNF1B and 14 neighboring genes. This locus is a hot spot for mutation since it is flanked by segmental repeats that can undergo homologous recombination during meiosis resulting in deletion of the intervening 1.4 Mb region.32 In addition to kidney abnormalities and diabetes caused by loss of HNF1B, individuals with 17q12 deletions often exhibit neurodevelopmental disorders, including autism spectrum disorder and attention-deficit/hyperactivity disorder, and facial dysmorphism.25; 32; 33 The neurological phenotypes are generally not seen in individuals with intragenic HNF1B deletions, suggesting that other genes within the deleted segment are responsible. One such candidate gene, LHX1, encodes a transcription factor that is involved in brain development and is also a downstream target of HNF1B32 The renal failure in 17q12 deletion individuals is less severe than those with an intragenic mutation,25; 30 possibly due to compensatory alterations in DNA methylation secondary to the 1,4-Mb deletion.25

In contrast to humans, mice that are heterozygous for an Hnf1b-null allele do not develop kidney cysts or other abnormalities, at least over the period of observation.6 The reason for this discrepancy is unclear, although a similar phenomenon has been described for mutations of Pkd1 and Pkd2. Various mechanisms may explain how heterozygous germline mutations produce a mutant phenotype despite the presence of a wild-type allele, including loss of heterozygosity (“second hit”), haploinsufficiency, and dominant-negative mutations. Kidneys from humans who are heterozygous for HNF1B mutations show preserved expression of the wild-type allele, arguing against a second hit mechanism.34 Some intragenic mutations are predicted to encode a mutant protein with an altered transcriptional activation domain but intact dimerization and DNA-binding domains. When co-expressed with wild-type HNF-1β in cell assays, such variants interfere with DNA-binding or transactivation and function as dominant-negative mutants.6 For other HNF1B mutations, haploinsufficiency may be responsible for the mutant phenotype.

2.2. Renal cysts and diabetes

Mutations of HNF1B in humans produce maturity-onset diabetes of the young, type 5 (MODY5). MODY is a monogenic disorder characterized by autosomal dominant inheritance of early-onset, non-autoimmune diabetes mellitus.35 Most affected individuals are diagnosed before 25 years of age, lack islet autoantibodies, and are neither obese nor insulin-dependent. MODY is genetically heterogenous and can arise from mutations in at least 14 genes. Mutations of glucokinase (GCK) and HNF1A are responsible for most cases.36 Mutations of HNF1B cause <5% of MODY and usually produce a more severe diabetic phenotype that often requires insulin replacement therapy. Nishigori et al. first reported the association of MODY5 with non-diabetic kidney disease presenting with renal dysfunction, hypertension, absent or low-grade proteinuria, and bilateral cystic kidneys.37 Subsequent studies have confirmed this association and additionally found that the renal phenotype is more penetrant than diabetes, so the absence of diabetes does not exclude the diagnosis.

Bingham et al. coined the term renal cysts and diabetes (RCAD) to describes patients with renal cysts and maturity-onset diabetes of the young caused by mutations of HNF1B38 Mutations can occur throughout the gene,26 although they are most common in exons 1-5. Of these mutations, the ones that occur in exons 2-4 affect the DNA binding domain.38 Cysts can be detected in utero.38 Histological analysis reveal kidney cysts, renal dysplasia, and oligomeganephronia. There is also considerable variation in kidney size in RCAD patients, such that kidneys are enlarged in fetuses and newborns and hypoplastic in adults. This size difference is thought to be due to growth cessation of the kidney after childhood.39 Most patients have impaired renal function, but the magnitude of renal failure varies among individuals with identical mutations.38

In addition to diabetes and kidney cysts, manifestations of RCAD may include hyperuricemia, gout, genital-tract malformations,38; 40 and hypomagnesemia.41 HNF-1β is required for pancreas development, and diabetes is thought to be due to reduced insulin secretion as a result of pancreatic hypoplasia.38 Hyperuricemia and hypomagnesemia arise from decreased expression of UMOD, organic anion transporters, and FXYD2, which regulate uric acid secretion and magnesium reabsorption in the nephron. HNF1B is expressed in the Müllerian duct, and HNF1B mutations are associated with female genital tract malformations such as bicornuate uterus, uterus didelphys, and vaginal aplasia. Hyperparathyroidism due to derepression of PTH and hypokalemia, which may be due to hypomagnesemia and down-regulation of KCNJ10 and SLC12A1, have also been described.42; 43

2.3. Multicystic dysplastic kidneys

Consistent with the essential role of HNF-1β in kidney development in mice, mutations of human HNF1B are causally associated with renal developmental abnormalities, some of which present as cystic kidneys. Mutations of HNF1B account for 10-30% of cases of congenital anomalies of the kidney and urinary tract (CAKUT).44 CAKUT includes renal agenesis, hypoplasia, dysplasia, hydronephrosis, horseshoe kidney, and multicystic dysplastic kidney (MCDK), all of which have been observed in humans with HNF1B mutations.45; 46 In several large pediatric series, mutations of HNF1B were among the most common genetic causes of renal hypodysplasia.4749 Kidney function can range from normal to ESRD depending on the severity of kidney abnormalities and whether they are unilateral or bilateral.

MCDK is a relatively common renal developmental abnormality in which normal kidney structure is replaced by clusters of non-communicating cysts.46 MCDK can be inherited or sporadic, unilateral or bilateral, and may be associated with ipsilateral urinary tract abnormalities, such as ureteral atresia. Histology shows disorganized kidney architecture, loss of corticomedullary boundary, primitive tubules surrounded by fibromuscular collars, and nests of metaplastic cartilage.50 MCDK has a frequency of 1 in 1,000 to 4,300 live births,46; 51 and one in 10 patients with unilateral MCDK have HNF1B mutations.52 A recent report describing a large autopsy series of fetuses carrying HNF1B mutations showed that most had unilateral or bilateral enlarged cystic kidneys with characteristic histological features of renal dysplasia.53 Urinary tract abnormalities were seen in 25% of cases.

2.4. Glomerulocystic kidney disease

GCKD is a rare autosomally-inherited disorder characterized by cortical glomerular cysts that can lead to chronic renal failure. Kidney histology shows cystic dilation of Bowman’s capsule with atrophic glomerular tufts present in at least 5% of the cysts.54 Concomitant tubular cysts may be present in the renal cortex or medulla but suggest alternative diagnoses such as ADPKD or tuberous sclerosis. GCKD is most common in neonates and infants with a family history of ADPKD;55 however, linkage studies found that some affected individuals do not have PKD1 or PKD2 mutations.55; 56 Germline mutations in HNF1B were found in some families with the hypoplastic form of GCKD.56 Individuals with HNF1B-related GCKD present with hypoplastic kidneys with glomerular cysts, malformations in the calyces and collecting systems, and early-onset diabetes.26; 56; 57

The mechanism of glomerular cystogenesis is not known, but primary cilia, Wnt signaling, proximal tubule malformation, and urinary tract obstruction have been implicated.54 HNF-1β is expressed in the parietal epithelium of Bowman’s capsule suggesting that the formation of glomerular cysts is cell autonomous. Glomerular cysts are also seen in Glis3 and TAZ/Wwtr1 knockout mice, and Jetten et al. have shown that cilia-localized Glis3 interacts with TAZ/Wwtr1 suggesting that they function in a common pathway.58 Wwtr1, which is an effector in the Hippo signaling pathway, is a downstream target of HNF-1β (see below), raising the possibility that glomerular cysts in HNF1B mutants arise from down-regulation of Wwtr1. Recent studies have shown that deletion of the transcription factor ZEB2 in mice produces glomerular cysts,59 and our studies have shown that HNF1b regulates ZEB2 expression. However, expression of ZEB2 is elevated in HNF-1β mutant kidneys rather than decreased, so the role of ZEB2 in HNF1B-related GCKD remains uncertain.

2.5. Autosomal dominant tubulointerstitial kidney disease

Autosomal dominant tubulointerstitial kidney disease (ADTKD) is a recently recognized monogenic disorder characterized by progressive renal insufficiency, normal or small kidney size, bland urinary sediment, and absence of nephrotoxin exposure or other known causes of CKD.40 ADTKD includes the diseases previously known as medullary cystic kidney disease (MCKD) and familial juvenile hyperuricemic nephropathy (FJHN). Affected individuals present with elevated serum creatinine, absent or low-grade proteinuria, impaired urinary concentration, and frequently hyperuricemia. Renal biopsy is rarely performed, but kidney histology shows interstitial fibrosis, tubular atrophy, tubular microcysts or macrocysts, basement membrane abnormalities, and absence of immune complex deposition. ESRD can develop over a wide age range from 17 to 75 years with a mean age of 47 years. ADTKD can arise from mutations in UMOD encoding uromodulin (Tamm-Horsfall protein), REN encoding renin, MUC1 encoding mucin-1, SEC61A1 encoding the Sec61 translocon, DNAJ11 encoding the chaperone protein ERdj3, and HNF1B60 Several of these genes are involved in proteostasis, and mutations result in accumulation of misfolded proteins, endoplasmic reticulum stress, induction of the unfolded protein response, and renal fibrosis.61

HNF1B-associated ADTKD (ADTKD-HNF1B), also known as HNF1B nephropathy, can be difficult to diagnose because the manifestations are non-specific and vary within affected families. Individuals with ADTKD-HNF1B may have multiple renal cysts and other abnormalities associated with CAKUT.62,63 Unlike other forms of ADTKD, ADTKD-HNF1B can present with extrarenal abnormalities including diabetes, elevated liver enzymes, autism spectrum disorder, cognitive impairment, pancreatic hypoplasia, female genital tract malformations, and hypomagnesemia.63; 64 These extrarenal manifestations can assist with diagnosis of HNF1B-related ADTKD. Faguer et al. developed an HNF1B score system based on the major clinical phenotypes as a method of pre-screening individuals for genetic testing.65 The score is calculated based on the occurrence of family history, renal, liver, pancreas and genital tract abnormalities.

2.6. Polycystic kidney disease

Although mutations of HNF1B do not cause typical PKD, there is a strong association between HNF-1β and both the dominant and recessive forms of PKD. As described in greater detail below, HNF-1β directly regulates the transcription of PKD2 and PKHD1, genes that cause ADPKD and ARPKD, respectively. Genetic studies using HNF-1β knockout mice and detailed molecular characterization of HNF-1β functions in kidney epithelial cells have shown that mutations of HNF-1β phenocopy many characteristics of PKD.66 The cystic kidney disease observed in humans with HNF1B mutations resembles ADPKD, and HNF1B mutations and have been detected in some individuals who were initially diagnosed with ADPKD but were not linked to PKD1 or PKD2,67; 68 The presence of atypical features such as unilateral disease, very early-onset, or extrarenal manifestations may assist with the correct diagnosis. Some phenotypic overlap between ADTKD and ADPKD also exists. Mutations of UMOD, MUC1, SEC61A1, DNAJB11, and HNF1B, which cause ADTKD, can also produce kidney cysts that phenocopy early-onset ADPKD.67; 68 The reason why mutations of these genes can present with primarily cystic kidney disease or primarily tubulointerstitial fibrosis is not clear, but may relate to developmental timing.

Expression of HNF-1β is reduced in cystic kidneys from individuals with ADPKD. Song et al. performed RNA expression profiling of individual kidney cysts derived from humans with ADPKD due to mutations of PKD1 and found consistently reduced expression of HNF1B compared to normal kidney.69 Corresponding reductions in expression of 9 of 15 HNF-1β-dependent target genes and derepression of SOCS3 were observed. These results suggest that HNF-1β is downstream to PKD1. Although the molecular mechanism is not known, the decreased expression of HNF-1β may contribute to the PKD1 cystic phenotype.

Consistent with the latter, HNF1B has been identified as a candidate modifier gene in human ADPKD. As described elsewhere in this series, ADPKD shows highly variable intrafamilial and interfamilial expressivity, which has been attributed to environmental factors and the effect of modifier genes. A small percentage of individuals with PKD1 or PKD2 mutations display an early onset of ADPKD that closely resembles autosomal recessive polycystic kidney disease (ARPKD) and can result in perinatal and neonatal death. Bergmann et al. genotyped families with early-onset ADPKD and found four individuals from three families who had inherited a PKD1 mutation from one parent and an HNF1B mutation from the other parent.70 The occurrence of an HNF1B mutation together with a PKD1 mutation produced more severe, earlier-onset disease compared to either parent alone. Children or fetuses with both mutations presented with oligohydramnios, renal insufficiency, or early-onset ESRD, and renal ultrasound showed bilateral enlarged echogenic or cystic kidneys. These results suggest that mutations of HNF1B may contribute to the phenotypic variation seen in humans with ADPKD.

3. Identification of genes regulated by HNF-1β

3.1. Mouse models of HNF-1β-related kidney diseases

To gain insights into the molecular pathogenesis of HNF1B-related kidney diseases, our laboratory and others have generated mouse models. HNF-1β is essential for visceral endoderm formation, and constitutive ablation of HNF-1β in the mouse is embryonic lethal.6; 71 Therefore, kidney-specific transgenics and gene targeting were required. In the first such approach, we generated transgenic mice expressing a dominant-negative HNF-1β (DN-HNF-1β) mutant lacking the carboxy-terminal transactivation domain under the control of a kidney-specific cadherin-16 promoter.72 DN-HNF-1β transgenic mice develop glomerular and tubular cysts and ESRD, which recapitulates the phenotype of humans with HNF1B mutations. Analysis of candidate genes showed marked reduction of Pkhd1 mRNA transcripts in the cyst epithelium compared to wide-type tubules. Pkhd1 encodes fibrocystin or polyductin, a transmembrane protein localized in cilia, and is the most frequently mutated gene in autosomal recessive polycystic kidney disease (ARPKD).73 The function of fibrocystin is not known, but its expression pattern closely overlaps with HNF-1β. Biochemical studies showed that transcription of Pkhd1 is directly regulated by HNF-1β in renal collecting duct cells.

Kidney-specific ablation of HNF-1β in renal tubules using Cre/loxP recombination also results in the formation of kidney cysts, confirming the results seen in DN-HNF-1β transgenic mice.74 In addition to decreased expression of Pkhd1, the expression of cystic disease genes Pkd2, Umod, Ift88, and Nphp1 is also downregulated. Chromatin immunoprecipitation showed the presence of HNF-1β binding sites in the Pkhd1, Umod, Pkd2, and Ift88 genes, suggesting that the transcription of these genes is directly regulated by HNF-1β.

Interestingly, the effects of HNF-1β inactivation are dependent on cell proliferation.13 In proliferating cells, loss of HNF-1β inhibits transcription of downstream target genes, whereas expression of target genes is unaffected when HNF-1β is inactivated in quiescent cells. This property may be related to the bookmarking function described earlier and suggests that HNF-1β binding to mitotic chromosomes is required to maintain the expression of target genes in daughter cells. Consequently, inactivation of HNF-1β in mice at postnatal day 1 when tubules are rapidly proliferating results in decreased expression of Pkd2 and Pkhd1 and the formation of kidney cysts. In contrast, inactivation of HNF-1β in mice after 10 days of age, when tubules are more quiescent, does not affect Pkd2 or Pkhd1 expression and does not produce cysts for up to 6 months. When adult mutant mice are subjected to acute kidney injury, cell proliferation and cyst formation are stimulated. Similar findings have been made in Kif3a and Pkd1 knockout mice, which suggests that abnormal repair after kidney injury may be a common factor in cyst initiation.75 Differences in kidney injury and cell proliferation may contribute to the phenotypic variability of HNF1B mutations.

3.2. Regulation of PKD-relevant genes

To identify additional gene networks regulated by HNF-1β, we performed genome-wide chromatin immunoprecipitation (ChIP-chip and ChIP-seq) in cultured renal epithelial cells.76; 77 By combining these approaches with RNA transcriptome profiling (microarrays and RNA-seq), we identified genes that are dysregulated in HNF-1β mutant cells, contain nearby HNF-1β binding sites, and are likely to represent direct transcriptional targets of HNF-1β.74; 77 These studies revealed that HNF-1β regulates the expression of many genes that are associated with cystic kidney disease in humans and rodents, including Pkhd1 (ARPKD); Pkd2 (ADPKD); Umod (ADTKD, MCKD); Cystin (cpk mouse); Nphp1, Nphp3, and Glis2 (nephronophthisis), Bicc1 (jcpk mouse); Kif12 (cpk modifier gene); and Wwtr1 (TAZ) (74; 7680 and unpublished data). The above studies reveal that HNF-1β regulates a transcriptional network containing multiple cystic disease genes. Loss-of-function mutations of HNF1B decrease the expression of cystic disease genes, which leads to cyst formation.

The HNF-1β-regulated gene Kif12 was first identified as a candidate modifier gene in the congenital polycystic kidney (cpk) mouse model of polycystic kidney disease.81 We demonstrated that mutations of HNF-1β inhibit Kif12 transcription by altering histone modifications and coregulatory recruitment at its promoter region.82 As Kinesin-12 family members are involved in orienting cell division,83 downregulation of Kif12 may underlie the abnormal planar cell polarity observed in HNF1B mutants (see below). In addition to transcriptional activation, HNF-1β can also function as a transcriptional repressor. In this case, mutations of HNF1B lead to increased target gene expression. One example is suppressor of cytokine signaling 3 (SOCS3), which is upregulated in HNF-1β mutant kidneys. SOCS3 inhibits HGF-induced tubulogenesis by reducing phosphorylation of Erk and STAT-3.76 Interestingly, Socs3 mRNA is highly expressed in human polycystic kidneys in which HNF-1β expression is reduced,69 and PKD1 mutant cells show activation of JAK2-STAT3 pathway with increased SOCS3 expression.84 These findings suggest that upregulation of SOCS3 may be a common feature of cystic kidney diseases.

4. Regulation of PKD signaling pathways

Polycystic kidney disease is characterized by dysregulation of numerous intracellular and intercellular signaling pathways. Analysis of HNF-1β mutant cells and kidneys has revealed dysregulation of many of the same PKD pathways. In this section, we review the role of HNF-1β in signaling pathways that are disrupted in cystic kidney diseases, focusing on cAMP signaling, Wnt signaling, planar cell polarity, and renal fibrosis.

4.1. cAMP signaling

Activation of cAMP signaling is a hallmark of PKD and forms the basis for treatment with tolvaptan and other drugs that reduce intracellular cAMP levels.85 Cyclic AMP levels are governed by the activities of G-protein coupled receptors, adenylyl cyclases, and cAMP phosphodiesterases.86 Cyclic AMP levels are elevated in many animal models of PKD, including Hnf1b mutant mice;87 however, the mechanism responsible for increased cAMP is poorly understood. We have shown that one protein complex in the primary cilium comprises adenylyl cyclase 5/6 (AC5/6), a scaffolding protein A-kinase anchoring protein 150 (AKAP150), and protein kinase A.87 Genetic mutations of Pkd2 and Hnf1b in mice result in increased cAMP levels that are dependent on activation of AC5/6. Polycystin-2 interacts with the AC5/6 complex through its carboxy-terminus and thereby regulates cAMP levels, possibly by allowing Ca2+ entry and inhibiting the Ca2+-sensitive AC5/6 isoforms. Loss of polycystin-2 would relieve this inhibition resulting in increased cAMP levels. Consistent with this model, genetic ablation of AC5 reduces cAMP levels and cyst growth in Pkd2 mutant mice.88 Since expression of polycystin-2 is controlled by HNF-1β, this mechanism could also explain the increased cAMP levels in HNF1B mutants.

In addition, the increased cAMP levels in HNF-1β mutant cells and kidneys may reflect transcriptional downregulation of a cAMP-specific member of the phosphodiesterase family, PDE4C.87 Phosphodiesterases mediate the conversion of cAMP to AMP and thereby terminate cAMP signaling.89 We found that PDE4C is located in the primary cilia of renal epithelial cells where it interacts with the AC5/6-AKAP150-PC2 complex.87 ChIP-ChIP showed that HNF-1β binds to the proximal promoter of Pde4c and is required for its transcription.87 Decreased expression of PDE4C in HNF1B mutant cells would result in reduced catabolism of cAMP and elevated cAMP levels. Collectively, these studies reveal novel mechanisms for dysregulation of cAMP signaling in cystic kidney diseases through down-regulation of Pkd2 and Pde4c. Inhibition of AC5 or stimulation of PDE4C may represent alternative approaches to decrease cAMP levels in cystic kidney diseases.

4.2. Canonical Wnt signaling

Wnts are secreted glycoproteins that play critical roles in many biological processes including embryonic development, cell fate determination, tissue homeostasis, and cell survival.90 Deregulation of Wnt signaling is frequently seen in human diseases such as cancer.91 The Wnt signaling pathway can be classified into canonical Wnt/β-catenin dependent and non-canonical/β-catenin independent. Canonical Wnt signaling is activated by binding of Wnt ligands to Frizzled (Fzd) and LDL-related protein (LRP) cell surface receptors.92 Ligand binding results in the membrane sequestration of a multiprotein complex containing APC and GSK3β, which normally phosphorylates β-catenin and targets it for proteasomal degradation.93 Sequestration of the destruction complex permits cytosolic accumulation of β-catenin, which translocates to the nucleus where it interacts with TCF/LEF transcription factors at Wnt Responsive Elements (WRE) to activate Wnt target gene expression.94

Several studies suggest that canonical Wnt signaling plays an important role in cystic kidney disease. For example, genetic ablation of kinesin subunit II (Kif3a) in mice produces cystic kidneys with the upregulation of β-catenin in cyst epithelium.95 Conditional inactivation of Ape, a scaffold protein contained in the cytosolic β-catenin destruction complex, produces kidney cysts lined by a hyperproliferative epithelium.96 Upregulation of GSK3β expression is associated with cyst expansion in mice and humans with PKD.97 The extracellular domain of polycystin-1 has been shown to bind to Wnt ligands and mediate Ca2+ influx through heterodimer complex formation with polycystin-2.98 In addition, the cytosolic carboxy-terminal domain of PC1 binds to β-catenin and downregulates canonical Wnt signaling.99 Conversely, mutation of Pkd2 increases β-catenin signaling in kidney tubules and results in cyst expansion, which can be prevented by Wnt inhibitors.100

Transcriptomic analysis showed that canonical Wnt signaling is highly activated in HNF-1β knockout cells.79; 101 Ablation of HNF-1β in mIMCD3 renal epithelial cells produces hyperresponsiveness to exogenous Wnt ligands and increases expression of Wnt target genes, including Axin2, Ccdc80, and Rnf43. Levels of β-catenin and expression of Wnt target genes are also increased in HNF-1β mutant mouse kidneys. In agreement with the mouse model, humans with HNF1B mutations display bilateral enlarged multicystic kidneys with accumulation of β-catenin in the hyperplastic cyst-lining cells.34 Genome-wide chromatin immunoprecipitation sequencing (ChIP-seq) in wild-type and mutant cells showed that ablation of HNF-1β produces a 6-fold increase in the number of genomic sites that are occupied by β-catenin.101 Remarkably, 50% of the sites that are occupied by β-catenin in HNF-1β mutant cells colocalize with HNF-1β-occupied sites in wild-type cells. The striking association of β-catenin and HNF-1β on WREs suggests a direct and widespread transcriptional repression of canonical Wnt signaling by HNF-1β. We further demonstrated that HNF-1β directly competes with the binding of β-catenin/LEF complexes to WREs through a novel composite motif containing a TCF/LEF binding site and an overlapping HNF-1β half-site. This study reveals a novel mechanism whereby a transcription factor constrains canonical Wnt activation by competing with β-catenin/LEF chromatin binding. Hyperactivation of canonical Wnt signaling due to loss of HNF-1β may contribute to cyst initiation and expansion in cystic kidney disease.

4.3. Planar cell polarity

During normal kidney organogenesis, developing renal tubules elongate without increasing their diameter. Fischer et al. showed that the mitotic spindles in dividing tubular cells are oriented parallel to the tubule axis.102 Consequently, cell division produces daughter cells that are aligned longitudinally, which results in tubule elongation without tubular dilation. Karner et al. found evidence for convergent extension in the developing mouse kidney, such that cell intercalation results in tubular elongation and a decrease in the number of cells in tubular cross sections.103 Both oriented cell division and convergent extension are manifestations of planar cell polarity (PCP). In HNF-1β mutant kidneys, the mitotic spindles in dividing tubular cells are no longer oriented parallel to the tubular axis.102 These data indicate that loss of HNF-1β disrupts PCP, which may contribute to tubular dilation and cyst formation. Disruption of PCP is also seen in a rat model of PKD caused by mutations of Pkhd1 (pck rat).102 In contrast, abnormalities in OCD were not observed in pre-cystic tubules in Pkd1 and Pkd2 mutant mice, indicating that early disruption of PCP is not a universal finding in PKD.104

PCP is regulated by non-canonical (β-catenin-independent) Wnt signaling in which binding of Wnt ligands to Fzd receptors results in activation of RhoA kinase.74 Studies by Karner et al. suggest that Wnt9b signaling through the non-canonical pathway may be involved in the establishment of PCP in the developing kidney, and mice homozygous for a hypomorphic Wnt9b allele form kidney cysts.103 In developing Wnt9b mutant tubules, cell elongation occurs in random orientation and the number of cells in tubular cross sections does not decrease as in wild-type kidneys, suggesting a defect in convergent extension. Consistent with inhibition of the non-canonical Wnt pathway, the phosphorylation of downstream RhoA is decreased. Since HNF-1β directly regulates Wnt9b transcription (above), this mechanism may also underlie the PCP defects in HNF-1β mutant kidneys. In addition, ChIP-seq studies have shown that HNF-1β binds to genes encoding core PCP proteins, such as Pricklel.76; 79

4.4. Renal fibrosis

Kidney fibrosis is a common final pathway in many forms of chronic kidney disease including cystic kidney diseases. Humans with HNF1B mutations can present with ADTKD, in which renal interstitial fibrosis is prominent. HNF-1β mutant mice produced using a collecting duct-specific Cre deletion develop more slowly progressive cystic kidney disease associated with significant renal fibrosis.2 To unravel the mechanism whereby loss of HNF-1β in renal epithelial cells produces interstitial fibrosis, we generated HNF-1β-deficient mIMCD3 cells using CRISPR-based gene editing.105 Characterization of the cells revealed that loss of HNF-1β induces epithelial-mesenchymal transition (EMT), as evidenced by spindle-shaped cell morphology, increased cell motility and invasiveness, and expression of mesenchymal markers.79 EMT is also observed in vivo in HNF-1β mutant mice, but genetic lineage analysis showed that EMT does not directly contribute to the formation of renal interstitial fibroblasts. Instead, we identified Twst2 as a direct target of HNF-1β that regulates an EMT transcriptional network in renal epithelial cells. Upregulation of Twist2 increases the expression of EMT transcription factors such as SnaiH, Snail2, Zeb1 and Zeb2, which in turn induces aberrant TGF-β signaling and fibroblast activation within the kidney interstitium. Snai1 also represses Hnf1b itself, suggesting that a positive feed-forward loop may promote fibrosis.105 The EMT mechanisms discovered in HNF-1β mutant cell and kidneys may underlie the pathophysiology of renal fibrosis in ADTKD and other cystic kidney diseases.

5. Regulation of noncoding RNAs

In addition to regulating the transcription of protein-coding genes, HNF-1β also regulates the expression of noncoding RNAs. As discussed in greater detail elsewhere in this series, much of the human genome is transcribed into RNA species that are not directly translated into proteins but instead have regulatory functions. Such noncoding RNAs include microRNAs (miRNA), long noncoding RNAs (IncRNA), enhancer RNAs (eRNA), circular RNAs (circRNA), and others. Microarray and RNA-seq experiments revealed that the expression of noncoding RNAs is dysregulated in HNF-1β mutant cells, and in some cases HNF-1β directly regulates the transcription of noncoding RNAs.

The first such noncoding RNA identified was a previously unannotated IncRNA encoding the miR-200 microRNA cluster.106 Long noncoding RNAs are >200 nucleotides in length and do not contain a long open-reading frame. LncRNAs are epigenetic regulators that control recruitment of transcription factors and chromatin remodeling complexes, RNA editing and splicing, sequestration of miRNAs, and protein translation.107 MicroRNA microarray analysis of renal epithelial cells expressing dominant-negative mutant HNF-1β revealed decreased expression of the miR-200b/200a/429 cluster. This microRNA cluster is of interest, since its expression is epithelial-specific, similar to HNF-1β, and down-regulation of miR-200 induces epithelial-mesenchymal transition, in part through disinhibition of the EMT transcription factors ZEB1 and ZEB2. Unexpectedly, we found that in renal epithelial cells, the miR-200b/a/429 cluster is encoded by a 28-kb IncRNA, the transcription of which is under the direct control of HNF-1β.106 Ablation of HNF-1β results in decreased IncRNA and miR-200 expression, which leads to increased levels of downstream targets, including ZEB2 and PKD1108 Re-expression of miR-200 reversed these abnormalities. These studies identified a novel mechanism for HNF-1β-dependent epigenetic regulation of PKD1 and revealed an additional pathway whereby loss of HNF-1β leads to EMT and renal fibrosis.

Some noncoding RNAs show increased expression following ablation of HNF-1β. Among these is the miR-17~92 cluster, overexpression of which is linked to PKD.109 Expression of miR-17~92 is upregulated in HNF-1β mutant kidneys and other models of human and mouse PKD. In contrast to miR-200, the expression of miR17~92 does not appear to be directly regulated by HNF-1β. Rather, the transcription of miR-17~92 is regulated by c-Myc.110

6. Conclusions

HNF-1β is an essential transcription factor that regulates gene expression during kidney development. Human cystic kidney diseases such as RCAD, MCDK, GCKD, and ADTKD are associated with mutations in HNF1B. Mutations in HNF1B can be familial or de novo and produce a broad range of phenotypes. HNF-1β directly regulates the expression of PKHD1, PKD2, and other genes involved in cystic kidney diseases (Figure 3a). HNF-1β also regulates cAMP levels in ADPKD through PDE4C (Figure 3b). Both canonical and non-canonical Wnt signaling pathways play a role in cystogenesis and are regulated by HNF-1β (Figure 3c). HNF-1β inhibits EMT by inhibiting TWIST2 (Figure 3d). HNF-1β regulation of noncoding RNAs also inhibits EMT (Figure 3e). Studies on HNF-1β-regulated signaling pathways have provided general insights into the mechanism of cyst formation and identified potential targets for therapeutic intervention in human cystic kidney diseases.

Figure 3. Roles of HNF-1β in renal cystogenesis.

Figure 3.

Schematic diagram showing the central role of HNF-1β in the transcriptional and post-transcriptional regulation of multiple pathways involved in renal cystogenesis, including a) Expression of cystic kidney disease genes; b) cAMP signaling; c) Wnt signaling; d) Renal fibrosis; and e) Noncoding RNAs. Mutations of HNF-1β produce kidney cysts through down-regulation of PKD2, PKHD1, and other cystic disease genes; down-regulation of PDE4C leading to increased cAMP; increased β-catenin/LEF binding leading to upregulation of canonical Wnt signaling and increased c-Myc and miR-17; and upregulation of Twist2 and down-regulation of miR-200 leading to EMT and renal fibrosis.

Figure 2. PKD-related signaling pathways.

Figure 2.

Signaling pathways that are regulated by HNF-1β and perturbed in PKD. a) Regulation of cAMP levels. HNF-1β regulates the expression of polycystin-2 (PC2) and phosphodiesterase 4C (PDE4C). PC2 inhibits calcium-sensitive adenyl cyclase 5 (AC5), while PDE4C decreases cAMP levels by converting cAMP into AMP. b) Canonical and non-canonical Wnt signaling pathways. In the Wnt-on state of canonical Wnt signaling, β-catenin translocates to the nucleus and forms complexes with TCF/LEF that compete with HNF-1β for binding to Wnt target genes. Non-canonical Wnt signaling is regulated by Wnt9b in the kidney and plays a role in planar cell polarity, c) HNF-1β inhibits the expression of Twist2, thereby reducing downstream TGF-β signaling that drives renal interstitial fibrosis.

Highlights.

  • HNF-1β is a POU-homeodomain transcription factor that regulates branching morphogenesis and mesenchymal-to-epithelial transition in the developing kidney

  • Mutations of HNF-1β in humans produce cystic kidney diseases

  • HNF-1β plays a central role in renal cystogenesis through the transcriptional and posttranscriptional regulation of cystic disease genes

Acknowledgements

We thank Karam Aboudehen for critically reviewing the manuscript. We apologize to colleagues whose work we were unable to cite because of space constraints. Research from the authors’ laboratory was supported by the National Institute of Diabetes, Digestive and Kidney Diseases of the National Institutes of Health under award number R37DK042921.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Declarations of interests: None

References

  • 1.Barbacci E, Reber M, Ott MO, Breillat C, Huetz F, Cereghini S. 1999. Variant hepatocyte nuclear factor 1 is required for visceral endoderm specification. Development. 126(21):4795–4805. [DOI] [PubMed] [Google Scholar]
  • 2.Ferre S, Igarashi P. 2018. New insights into the role of hnf-1beta in kidney (patho)physiology. Pediatr Nephrol. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Casemayou A, Fournel A, Bagattin A, Schanstra J, Belliere J, Decramer S, Marsal D, Gillet M, Chassaing N, Huart A et al. 2017. Hepatocyte nuclear factor-1beta controls mitochondrial respiration in renal tubular cells. J Am Soc Nephrol. 28(11):3205–3217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Lu P, Rha GB, Chi YI. 2007. Structural basis of disease-causing mutations in hepatocyte nuclear factor 1beta. Biochemistry. 46(43):12071–12080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hiesberger T, Shao X, Gourley E, Reimann A, Pontoglio M, Igarashi P. 2005. Role of the hepatocyte nuclear factor-1beta (hnf-1beta) c-terminal domain in pkhd1 (arpkd) gene transcription and renal cystogenesis. J Biol Chem. 280(11):10578–10586. [DOI] [PubMed] [Google Scholar]
  • 6.Barbacci E, Chalkiadaki A, Masdeu C, Haumaitre C, Lokmane L, Loirat C, Cloarec S, Talianidis I, Bellanne-Chantelot C, Cereghini S. 2004. Hnf1b/tcf2 mutations impair transactivation potential through altered co-regulator recruitment. Human Molecular Genetics. 13(24):11. [DOI] [PubMed] [Google Scholar]
  • 7.Cereghini S. 1996. Liver-enriched transcription factors and hepatocyte differentiation. FASEB J. 10(2):267–282. [PubMed] [Google Scholar]
  • 8.Nicosia A, Monaci P, Tomei L, De Francesco R, Nuzzo M, Stunnenberg H, Cortese R. 1990. A myosin-like dimerization helix and an extra-large homeodomain are essential elements of the tripartite dna binding structure of lfb1. Cell. 61(7):1225–1236. [DOI] [PubMed] [Google Scholar]
  • 9.Mendel DB, Khavari PA, Conley PB, Graves MK, Hansen LP, Admon A, Crabtree GR. Characterization of a cofactor that regulates dimerization of a mammalian homeodomain protein. 254(5039):1762–1767. [DOI] [PubMed] [Google Scholar]
  • 10.Ceska TA, Lamers M, Monaci P, Nicosia A, Cortese R, Suck D. 1993. The x-ray structure of an atypical homeodomain present in the rat liver transcription factor lfb1/hnf1 and implications for dna binding. EMBO J. 12(5): 1805–1810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bürglin TR, Affolter M. 2016. Homeodomain proteins: An update. Chromosoma. 125(3):497–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Rey-Campos J, Chouard T, Yaniv M, Cereghini S. 1991. Vhnf1 is a homeoprotein that activates transcription and forms heterodimers with hnf1. EMBO J. 10(6):1445–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Verdeguer F, Le Corre S, Fischer E, Callens C, Garbay S, Doyen A, Igarashi P, Terzi F, Pontoglio M. 2010. A mitotic transcriptional switch in polycystic kidney disease. Nat Med. 16(1):106–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Lerner J, Bagattin A, Verdeguer F, Makinistoglu MP, Garbay S, Felix T, Heidet L, Pontoglio M. 2016. Human mutations affect the epigenetic/bookmarking function of hnf1b. Nucleic Acids Res. 44(17):8097–8111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Costantini F. 2006. Renal branching morphogenesis: Concepts, questions, and recent advances. Differentiation. 74(7):402–421. [DOI] [PubMed] [Google Scholar]
  • 16.Costantini F, Kopan R. 2010. Patterning a complex organ: Branching morphogenesis and nephron segmentation in kidney development. Dev Cell. 18(5):698–712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Vainio S, Lin Y. 2002. Coordinating early kidney development: Lessons from gene targeting. Nat Rev Genet. 3(7):533–543. [DOI] [PubMed] [Google Scholar]
  • 18.Dressler GR. 2006. The cellular basis of kidney development. Annu Rev Cell Dev Biol. 22:509–529. [DOI] [PubMed] [Google Scholar]
  • 19.Lokmane L, Heliot C, Garcia-Villalba P, Fabre M, Cereghini S. 2010. Vhnf1 functions in distinct regulatory circuits to control ureteric bud branching and early nephrogenesis. Development. 137(2):347–357. [DOI] [PubMed] [Google Scholar]
  • 20.Massa F, Garbay S, Bouvier R, Sugitani Y, Noda T, Gubler MC, Heidet L, Pontoglio M, Fischer E. 2013. Hepatocyte nuclear factor 1beta controls nephron tubular development. Development. 140(4):886–896. [DOI] [PubMed] [Google Scholar]
  • 21.Desgrange A, Heliot C, Skovorodkin I, Akram SU, Heikkila J, Ronkainen VP, Miinalainen I, Vainio SJ, Cereghini S. 2017. Hnf1b controls epithelial organization and cell polarity during ureteric bud branching and collecting duct morphogenesis Development. England: 2017. Published by The Company of Biologists Ltd; p. 4704–4719. [DOI] [PubMed] [Google Scholar]
  • 22.Narlis M, Grote D, Gaitan Y, Boualia SK, Bouchard M. 2007. Pax2 and pax8 regulate branching morphogenesis and nephron differentiation in the developing kidney. J am soc nephrol. United States. p. 1121–1129. [DOI] [PubMed] [Google Scholar]
  • 23.Majumdar A, Vainio S, Kispert A, McMahon J, McMahon AP. 2003. Wnt11 and ret/gdnf pathways cooperate in regulating ureteric branching during metanephric kidney development. Development. 130(14):3175–3185. [DOI] [PubMed] [Google Scholar]
  • 24.Carroll T, Park J, Hayashi S, Majumdar A, McMahon A. 2005. Wnt9b plays a central role in the regulation of mesenchymal to epithelial transitions underlying organogenesis of the mammalian urogenital system. Developmental Cell. 9(2):283–292. [DOI] [PubMed] [Google Scholar]
  • 25.Clissold RL, Ashfield B, Burrage J, Hannon E, Bingham C, Mill J, Hattersley A, Dempster EL. 2018. Genome-wide methylomic analysis in individuals with hnf1b intragenic mutation and 17q12 microdeletion. Clin Epigenetics. 10(1):97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Alvelos MI, Rodrigues M, Lobo L, Medeira A, Sousa AB, Simao C, Lemos MC. 2015. A novel mutation of the hnf1b gene associated with hypoplastic glomerulocystic kidney disease and neonatal renal failure: A case report and mutation update. Medicine (Baltimore). 94(7):e469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bellanne-Chantelot C, Clauin S, Chauveau D, Collin P, Daumont M, Douillard C, Dubois-Laforgue D, Dusselier L, Gautier JF, Jadoul M et al. 2005. Large genomic rearrangements in the hepatocyte nuclear factor-1beta (tcf2) gene are the most frequent cause of maturity-onset diabetes of the young type 5. Diabetes. United States. p. 3126–3132. [DOI] [PubMed] [Google Scholar]
  • 28.Heidet L, Decramer S, Pawtowski A, Moriniere V, Bandin F, Knebelmann B, Lebre AS, Faguer S, Guigonis V, Antignac C et al. 2010. Spectrum of hnf1b mutations in a large cohort of patients who harbor renal diseases. Clin J Am Soc Nephrol. 5(6):1079–1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Edghill EL, Bingham C, Ellard S, Hattersley AT. 2006. Mutations in hepatocyte nuclear factor-1beta and their related phenotypes. J med genet. England. p. 84–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Dubois-Laforgue D, Cornu E, Saint-Martin C, Coste J, Bellanne-Chantelot C, Timsit J. 2017. Diabetes, associated clinical spectrum, long-term prognosis, and genotype/phenotype correlations in 201 adult patients with hepatocyte nuclear factor 1b (hnf1b) molecular defects. Diabetes care. United States: 2017 by the American Diabetes Association. p. 1436–1443. [DOI] [PubMed] [Google Scholar]
  • 31.Okorn C, Goertz A, Vester U, Beck BB, Bergmann C, Habbig S, Konig J, Konrad M, Muller D, Oh J et al. 2019. Hnf1b nephropathy has a slow-progressive phenotype in childhood-with the exception of very early onset cases: Results of the german multicenter hnf1b childhood registry. Pediatr Nephrol. 34(6):1065–1075. [DOI] [PubMed] [Google Scholar]
  • 32.Moreno-De-Luca D, Mulle JG, Kaminsky EB, Sanders SJ, Myers SM, Adam MP, Pakula AT, Eisenhauer NJ, Uhas K, Weik L et al. 2010. Deletion 17q12 is a recurrent copy number variant that confers high risk of autism and schizophrenia. Am J Hum Genet. 87(5):618–630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Clissold RL, Shaw-Smith C, Turnpenny P, Bunce B, Bockenhauer D, Kerecuk L, Waller S, Bowman P, Ford T, Ellard S et al. 2016. Chromosome 17q12 microdeletions but not intragenic hnf1b mutations link developmental kidney disease and psychiatric disorder. Kidney Int. 90(1):203–211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Haumaitre C, Fabre M, Cormier S, Baumann C, Delezoide AL, Cereghini S. 2006. Severe pancreas hypoplasia and multicystic renal dysplasia in two human fetuses carrying novel hnf1beta/mody5 mutations. Hum Mol Genet. 15(15):2363–2375. [DOI] [PubMed] [Google Scholar]
  • 35.Horikawa Y, Iwasaki N, Hara M, Furuta H, Hinokio Y, Cockburn BN, Lindner T, Yamagata K, Ogata M, Tomonaga O et al. 1997. Mutation in hepatocyte nuclear factor-1 beta gene (tcf2) associated with mody. Nat Genet. 17(4):384–385. [DOI] [PubMed] [Google Scholar]
  • 36.Firdous P, Nissar K, Ali S, Ganai BA, Shabir U, Hassan T, Masoodi SR. 2018. Genetic testing of maturity-onset diabetes of the young current status and future perspectives. Front Endocrinol (Lausanne). 9:253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Nishigori H, Yamada S, Kohama T, Tomura H, Sho K, Horikawa Y, Bell GI, Takeuchi T, Takeda J. 1998. Frameshift mutation, a263fsinsgg, in the hepatocyte nuclear factor-1beta gene associated with diabetes and renal dysfunction. Diabetes. 47(8):1354–1355. [DOI] [PubMed] [Google Scholar]
  • 38.Bingham C, Hattersley AT. 2004. Renal cysts and diabetes syndrome resulting from mutations in hepatocyte nuclear factor-1beta. Nephrol dial transplant. England. p. 2703–2708. [DOI] [PubMed] [Google Scholar]
  • 39.Waller SC, Rees L, Woolf AS, Ellard S, Pearson ER, Hattersley AT, Bingham C. 2002. Severe hyperglycemia after renal transplantation in a pediatric patient with a mutation of the hepatocyte nuclear factor-1beta gene. Am J Kidney Dis. 40(6):1325–1330. [DOI] [PubMed] [Google Scholar]
  • 40.Eckardt KU, Alper SL, Antignac C, Bleyer AJ, Chauveau D, Dahan K, Deltas C, Hosking A, Kmoch S, Rampoldi L et al. 2015. Autosomal dominant tubulointerstitial kidney disease: Diagnosis, classification, and management--a kdigo consensus report. Kidney Int. 88(4):676–683. [DOI] [PubMed] [Google Scholar]
  • 41.Adalat S, Woolf AS, Johnstone KA, Wirsing A, Harries LW, Long DA, Hennekam RC, Ledermann SE, Rees L, van’t Hoff W et al. 2009. Hnf1b mutations associate with hypomagnesemia and renal magnesium wasting. J Am Soc Nephrol. 20(5): 1123–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ferre S, Bongers EM, Sonneveld R, Cornelissen EA, van der Vlag J, van Boekel GA, Wetzels JF, Hoenderop JG, Bindels RJ, Nijenhuis T. 2013. Early development of hyperparathyroidism due to loss of pth transcriptional repression in patients with hnf1beta mutations? J Clin Endocrinol Metab. 98(10):4089–4096. [DOI] [PubMed] [Google Scholar]
  • 43.Verhave JC, Bech AP, Wetzels JF, Nijenhuis T. 2016. Hepatocyte nuclear factor 1beta-associated kidney disease: More than renal cysts and diabetes. J Am Soc Nephrol. 27(2):345–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Raaijmakers A, Corveleyn A, Devriendt K, van Tienoven TP, Allegaert K, Van Dyck M, van den Heuvel L, Kuypers D, Claes K, Mekahli D et al. 2015. Criteria for hnf1b analysis in patients with congenital abnormalities of kidney and urinary tract. Nephrol Dial Transplant. 30(5):835–842. [DOI] [PubMed] [Google Scholar]
  • 45.Choi HA, Lee DJ, Shin SM, Lee YK, Ko SY, Park SW. 2016. The prenatal and postnatal incidence of congenital anomalies of the kidneys and urinary tract (cakut) detected by ultrasound. Childhood Kidney Diseases. 20:4. [Google Scholar]
  • 46.Kara A, Gurgoze MK, Aydin M, Koc ZP. 2018. Clinical features of children with multicystic dysplastic kidney. Pediatr Int. 60(8):750–754. [DOI] [PubMed] [Google Scholar]
  • 47.Sanna-Cherchi S, Kiryluk K, Burgess KE, Bodria M, Sampson MG, Hadley D, Nees SN, Verbitsky M, Perry BJ, Sterken R et al. 2012. Copy-number disorders are a common cause of congenital kidney malformations. Am J Hum Genet. 91(6):987–997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Thomas R, Sanna-Cherchi S, Warady BA, Furth SL, Kaskel FJ, Gharavi AG. 2011. Hnf1b and pax2 mutations are a common cause of renal hypodysplasia in the ckid cohort. Pediatr Nephrol. 26(6):897–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Weber S, Moriniere V, Knuppel T, Charbit M, Dusek J, Ghiggeri GM, Jankauskiene A, Mir S, Montini G, Peco-Antic A et al. 2006. Prevalence of mutations in renal developmental genes in children with renal hypodysplasia: Results of the escape study. J Am Soc Nephrol. 17(10):2864–2870. [DOI] [PubMed] [Google Scholar]
  • 50.Hains DS, Bates CM, Ingraham S, Schwaderer AL. 2009. Management and etiology of the unilateral multicystic dysplastic kidney: A review. Pediatr Nephrol. 24(2):233–241. [DOI] [PubMed] [Google Scholar]
  • 51.Feldenberg LR, Siegel NJ. 2000. Clinical course and outcome for children with multicystic dysplastic kidneys. Pediatr Nephrol. 14(12):1098–1101. [DOI] [PubMed] [Google Scholar]
  • 52.Nakayama M, Nozu K, Goto Y, Kamei K, Ito S, Sato H, Emi M, Nakanishi K, Tsuchiya S, Iijima K. 2010. Hnf1b alterations associated with congenital anomalies of the kidney and urinary tract. Pediatr Nephrol. 25(6):1073–1079. [DOI] [PubMed] [Google Scholar]
  • 53.Duval H, Michel-Calemard L, Gonzales M, Loget P, Beneteau C, Buenerd A, Joubert M, Denis-Musquer M, Clemenson A, Chesnais AL et al. 2016. Fetal anomalies associated with hnf1b mutations: Report of 20 autopsy cases. Prenat Diagn. 36(8):744–751. [DOI] [PubMed] [Google Scholar]
  • 54.Bissler JJ, Siroky BJ, Yin H. 2010. Glomerulocystic kidney disease. Pediatr Nephrol. 25(10):2049–2056; quiz 2056–2049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Sharp CK, Bergman SM, Stockwin JM, Robbin ML, Galliani C, Guay-Woodford LM. 1997. Dominantly transmitted glomerulocystic kidney disease: A distinct genetic entity. J Am Soc Nephrol. 8(1):77–84. [DOI] [PubMed] [Google Scholar]
  • 56.Bingham C, Bulman MP, Ellard S, Allen LI, Lipkin GW, Hoff WG, Woolf AS, Rizzoni G, Novelli G, Nicholls AJ et al. 2001. Mutations in the hepatocyte nuclear factor-1beta gene are associated with familial hypoplastic glomerulocystic kidney disease. Am J Hum Genet. 68(1):219–224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mache CJ, Preisegger KH, Kopp S, Ratschek M, Ring E. 2002. De novo hnf-1 beta gene mutation in familial hypoplastic glomerulocystic kidney disease. Pediatr Nephrol. 17(12):1021–1026. [DOI] [PubMed] [Google Scholar]
  • 58.Kang HS, Beak JY, Kim YS, Herbert R, Jetten AM. 2009. Glis3 is associated with primary cilia and wwtr1/taz and implicated in polycystic kidney disease. Mol Cell Biol. 29(10):2556–2569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Rasouly HM, Kumar S, Chan S, Pisarek-Horowitz A, Sharma R, Xi QJ, Nishizaki Y, Higashi Y, Salant DJ, Maas RL et al. 2016. Loss of zeb2 in mesenchyme-derived nephrons causes primary glomerulocystic disease. Kidney Int. 90(6):1262–1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ayasreh N, Miquel R, Matamala A, Ars E, Torra R. 2017. A review on autosomal dominant tubulointerstitial kidney disease. Nefrología (English Edition). 37:9. [DOI] [PubMed] [Google Scholar]
  • 61.Kemter E, Frohlich T, Arnold GJ, Wolf E, Wanke R. 2017. Mitochondrial dysregulation secondary to endoplasmic reticulum stress in autosomal dominant tubulointerstitial kidney disease - umod (adtkd-umod). Sci Rep. 7:42970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Bleyer AJ, Kidd K, Zivna M, Kmoch S. 2017. Autosomal dominant tubulointerstitial kidney disease. Adv Chronic Kidney Dis. 24(2):86–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Cormican S, Connaughton DM, Kennedy C, Murray S, Zivna M, Kmoch S, Fennelly NK, O’Kelly P, Benson KA, Conlon ET et al. 2019. Autosomal dominant tubulointerstitial kidney disease (adtkd) in ireland. Ren Fail. 41(1):832–841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Devuyst O, Olinger E, Weber S, Eckardt KU, Kmoch S, Rampoldi L, Bleyer AJ. 2019. Autosomal dominant tubulointerstitial kidney disease. Nat rev dis primers. England. p. 60. [DOI] [PubMed] [Google Scholar]
  • 65.Faguer S, Chassaing N, Bandin F, Prouheze C, Garnier A, Casemayou A, Huart A, Schanstra JP, Calvas P, Decramer S et al. 2014. The hnf1b score is a simple tool to select patients for hnf1b gene analysis. Kidney Int. 86(5):1007–1015. [DOI] [PubMed] [Google Scholar]
  • 66.Igarashi P, Shao X, McNally BT, Hiesberger T. 2005. Roles of hnf-1beta in kidney development and congenital cystic diseases. Kidney Int. 68(5):1944–1947. [DOI] [PubMed] [Google Scholar]
  • 67.Chebib FT, Perrone RD, Chapman AB, Dahl NK, Harris PC, Mrug M, Mustafa RA, Rastogi A, Watnick T, Yu ASL et al. 2018. A practical guide for treatment of rapidly progressive adpkd with tolvaptan. J Am Soc Nephrol. 29(10):2458–2470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Cornec-Le Gall E, Torres VE, Harris PC. 2018. Genetic complexity of autosomal dominant polycystic kidney and liver diseases. (1): 13–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Song X, Di Giovanni V, He N, Wang K, Ingram A, Rosenblum ND, Pei Y. 2009. Systems biology of autosomal dominant polycystic kidney disease (adpkd): Computational identification of gene expression pathways and integrated regulatory networks. Hum mol genet. England, p. 2328–2343. [DOI] [PubMed] [Google Scholar]
  • 70.Bergmann C, von Bothmer J, Ortiz Bruchle N, Venghaus A, Frank V, Fehrenbach H, Hampel T, Pape L, Buske A, Jonsson J et al. 2011. Mutations in multiple pkd genes may explain early and severe polycystic kidney disease. J Am Soc Nephrol. 22(11):2047–2056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Coffinier C, Barra J, Babinet C, Yaniv M. 1999. Expression of the vhnf1/hnf1β homeoprotein gene during mouse organogenesis. Mechanisms of Development. 89(1):211–213. [DOI] [PubMed] [Google Scholar]
  • 72.Hiesberger T, Bai Y, Shao X, McNally BT, Sinclair AM, Tian X, Somlo S, Igarashi P. 2004. Mutation of hepatocyte nuclear factor-1 beta inhibits Pkhd1 gene expression and produces renal cysts in mice. J Clin Invest. 113(6):814–825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Igarashi P, Somlo S. 2002. Genetics and pathogenesis of polycystic kidney disease. J Am Soc Nephrol. 13(9):2384–2398. [DOI] [PubMed] [Google Scholar]
  • 74.Gresh L, Fischer E, Reimann A, Tanguy M, Garbay S, Shao X, Hiesberger T, Fiette L, Igarashi P, Yaniv M et al. 2004. A transcriptional network in polycystic kidney disease. EMBO J. 23(7): 1657–1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Patel V, Li L, Cobo-Stark P, Shao X, Somlo S, Lin F, Igarashi P. 2008. Acute kidney injury and aberrant planar cell polarity induce cyst formation in mice lacking renal cilia. Hum Mol Genet. 17(11): 1578–1590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ma Z, Gong Y, Patel V, Karner CM, Fischer E, Hiesberger T, Carroll TJ, Pontoglio M, Igarashi P. 2007. Mutations of hnf-1beta inhibit epithelial morphogenesis through dysregulation of socs-3. Proc Natl Acad Sci U S A. 104(51):20386–20391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Aboudehen K, Kim MS, Mitsche M, Garland K, Anderson N, Noureddine L, Pontoglio M, Patel V, Xie Y, DeBose-Boyd R et al. 2016. Transcription factor hepatocyte nuclear factor-1beta regulates renal cholesterol metabolism. J Am Soc Nephrol. 27(8):2408–2421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kompatscher A, de Baaij JHF, Aboudehen K, Hoefnagels A, Igarashi P, Bindels RJM, Veenstra GJC, Hoenderop JGJ. 2017. Loss of transcriptional activation of the potassium channel kir5.1 by hnf1beta drives autosomal dominant tubulointerstitial kidney disease. Kidney Int. 92(5):1145–1156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Chan SC, Zhang Y, Shao A, Avdulov S, Herrera J, Aboudehen K, Pontoglio M, Igarashi P. 2018. Mechanism of fibrosis in hnf1b-related autosomal dominant tubulointerstitial kidney disease. J Am Soc Nephrol. 29(10):2493–2509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hou X, Mrug M, Yoder BK, Lefkowitz EJ, Kremmidiotis G, D’Eustachio P, Beier DR, Guay-Woodford LM. 2002. Cystin, a novel cilia-associated protein, is disrupted in the cpk mouse model of polycystic kidney disease. J Clin Invest. 109(4):533–540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Mrug M, Li R, Cui X, Schoeb TR, Churchill GA, Guay-Woodford LM. 2005. Kinesin family member 12 is a candidate polycystic kidney disease modifier in the cpk mouse. J Am Soc Nephrol. 16(4):905–916. [DOI] [PubMed] [Google Scholar]
  • 82.Gong Y, Ma Z, Patel V, Fischer E, Hiesberger T, Pontoglio M, Igarashi P. 2009. Hnf-1beta regulates transcription of the pkd modifier gene kif12. J Am Soc Nephrol. 20(1):41–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kee YS, Ren Y, Dorfman D, Iijima M, Firtel R, Iglesias PA, Robinson DN. 2012. A mechanosensory system governs myosin ii accumulation in dividing cells. Mol Biol Cell. 23(8):1510–1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Jing Y, Wu M, Zhang D, Chen D, Yang M, Mei S, He L, Gu J, Qi N, Fu L et al. 2018. Triptolide delays disease progression in an adult rat model of polycystic kidney disease through the jak2-stat3 pathway. Am J Physiol Renal Physiol. 315(3):F479–F486. [DOI] [PubMed] [Google Scholar]
  • 85.Bergmann C, Guay-Woodford LM, Harris PC, Horie S, Peters DJM, Torres VE. 2018. Polycystic kidney disease. Nat Rev Dis Primers. 4(1):50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Torres VE, Harris PC. 2014. Strategies targeting camp signaling in the treatment of polycystic kidney disease. J Am Soc Nephrol. 25(1): 18–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Choi YH, Suzuki A, Hajarnis S, Ma Z, Chapin HC, Caplan MJ, Pontoglio M, Somlo S, Igarashi P. 2011. Polycystin-2 and phosphodiesterase 4c are components of a ciliary a-kinase anchoring protein complex that is disrupted in cystic kidney diseases. Proc Natl Acad Sci USA. 108(26): 10679–10684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wang Q, Cobo-Stark P, Patel V, Somlo S, Han PL, Igarashi P. 2018. Adenylyl cyclase 5 deficiency reduces renal cyclic amp and cyst growth in an orthologous mouse model of polycystic kidney disease. Kidney Int. 93(2):403–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Francis SH, Blount MA, Corbin JD. 2011. Mammalian cyclic nucleotide phosphodiesterases: Molecular mechanisms and physiological functions. Physiol Rev. 91(2):651–690. [DOI] [PubMed] [Google Scholar]
  • 90.MacDonald BT, Tamai K, He X. 2009. Wnt/beta-catenin signaling: Components, mechanisms, and diseases. Dev Cell. 17(1):9–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Nusse R, Clevers H. 2017. Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 169(6):985–999. [DOI] [PubMed] [Google Scholar]
  • 92.Cong F, Schweizer L, Varmus H. 2004. Wnt signals across the plasma membrane to activate the beta-catenin pathway by forming oligomers containing its receptors, frizzled and Irp. Development. 131 (20):5103–5115. [DOI] [PubMed] [Google Scholar]
  • 93.Valenta T, Hausmann G, Basler K. 2012. The many faces and functions of beta-catenin. Emboj. 31 (12):2714–2736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Gammons M, Bienz M. 2018. Multiprotein complexes governing wnt signal transduction. Curr Opin Cell Biol. 51:42–49. [DOI] [PubMed] [Google Scholar]
  • 95.Lin F, Hiesberger T, Cordes K, Sinclair AM, Goldstein LS, Somlo S, Igarashi P. 2003. Kidney-specific inactivation of the kif3a subunit of kinesin-ii inhibits renal ciliogenesis and produces polycystic kidney disease. Proc Natl Acad Sci U S A. 100(9):5286–5291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Qian CN, Knol J, Igarashi P, Lin F, Zylstra U, Teh BT, Williams BO. 2005. Cystic renal neoplasia following conditional inactivation of apc in mouse renal tubular epithelium. J Biol Chem. 280(5):3938–3945. [DOI] [PubMed] [Google Scholar]
  • 97.Tao S, Kakade VR, Woodgett JR, Pandey P, Suderman ED, Rajagopal M, Rao R. 2015. Glycogen synthase kinase-3beta promotes cyst expansion in polycystic kidney disease. Kidney Int. 87(6):1164–1175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kim S, Nie H, Nesin V, Tran U, Outeda P, Bai CX, Keeling J, Maskey D, Watnick T, Wessely O et al. 2016. The polycystin complex mediates wnt/ca(2+) signalling. Nat Cell Biol. 18(7):752–764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Lal M, Song X, Pluznick JL, Di Giovanni V, Merrick DM, Rosenblum ND, Chauvet V, Gottardi CJ, Pei Y, Caplan MJ. 2008. Polycystin-1 c-terminal tail associates with beta-catenin and inhibits canonical wnt signaling. Hum Mol Genet. 17(20):3105–3117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Li A, Xu Y, Fan S, Meng J, Shen X, Xiao Q, Li Y, Zhang L, Zhang X, Wu G et al. 2018. Canonical wnt inhibitors ameliorate cystogenesis in a mouse ortholog of human adpkd. JCI Insight. 3(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Chan SC, Zhang Y, Pontoglio M, Igarashi P. 2019. Hepatocyte nuclear factor-1beta regulates wnt signaling through genome-wide competition with beta-catenin/lymphoid enhancer binding factor. Proc Natl Acad Sci U S A. 116(48):24133–24142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Fischer E, Legue E, Doyen A, Nato F, Nicolas JF, Torres V, Yaniv M, Pontoglio M. 2006. Defective planar cell polarity in polycystic kidney disease. Nat Genet. 38(1):21–23. [DOI] [PubMed] [Google Scholar]
  • 103.Karner CM, Chirumamilla R, Aoki S, Igarashi P, Wallingford JB, Carroll TJ. 2009. Wnt9b signaling regulates planar cell polarity and kidney tubule morphogenesis. Nat Genet. 41(7):793–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Nishio S, Tian X, Gallagher AR, Yu Z, Patel V, Igarashi P, Somlo S. 2010. Loss of oriented cell division does not initiate cyst formation. J Am Soc Nephrol. 21(2):295–302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Boutet A, De Frutos CA, Maxwell PH, Mayol MJ, Romero J, Nieto MA. 2006. Snail activation disrupts tissue homeostasis and induces fibrosis in the adult kidney. Embo j. 25(23):5603–5613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Hajarnis SS, Patel V, Aboudehen K, Attanasio M, Cobo-Stark P, Pontoglio M, Igarashi P. 2015. Transcription factor hepatocyte nuclear factor-1beta (hnf-1beta) regulates microrna-200 expression through a long noncoding rna. J Biol Chem. 290(41):24793–24805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Yang G, Lu X, Yuan L. 2014. Lncrna: A link between rna and cancer. Biochim Biophys Acta. 1839(11):1097–1109. [DOI] [PubMed] [Google Scholar]
  • 108.Patel V, Hajarnis S, Williams D, Hunter R, Huynh D, Igarashi P. 2012. Micrornas regulate renal tubule maturation through modulation of pkd1. J Am Soc Nephrol. 23(12):1941–1948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Patel V, Williams D, Hajarnis S, Hunter R, Pontoglio M, Somlo S, Igarashi P. 2013. Mir-17~92 mirna cluster promotes kidney cyst growth in polycystic kidney disease. Proc Natl Acad Sci U S A. 110(26):10765–10770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Hajarnis S, Lakhia R, Yheskel M, Williams D, Sorourian M, Liu X, Aboudehen K, Zhang S, Kersjes K, Galasso R et al. 2017. Microrna-17 family promotes polycystic kidney disease progression through modulation of mitochondrial metabolism. Nat Commun. 8:14395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Wang Y, Zhao Y, Zhang J, Yang Y, Liu F. 2017. A case of a novel mutation in hnf1beta-related maturity-onset diabetes of the young type 5 with diabetic kidney disease complication in a Chinese family. J diabetes complications. United States: 2016 Elsevier Inc p. 1243–1246. [DOI] [PubMed] [Google Scholar]

RESOURCES