Click here to close
Hello! We notice that you are using Internet Explorer, which is not supported by Xenbase and may cause the site to display incorrectly.
We suggest using a current version of Chrome,
FireFox, or Safari.
J Am Soc Nephrol
2023 Mar 01;343:412-432. doi: 10.1681/ASN.2022010076.
Show Gene links
Show Anatomy links
HNF1B Alters an Evolutionarily Conserved Nephrogenic Program of Target Genes.
Grand K
,
Stoltz M
,
Rizzo L
,
Röck R
,
Kaminski MM
,
Salinas G
,
Getwan M
,
Naert T
,
Pichler R
,
Lienkamp SS
.
???displayArticle.abstract???
SIGNIFICANCE STATEMENT: Mutations in hepatocyte nuclear factor-1 β ( HNF1B ) are the most common monogenic causes of congenital renal malformations. HNF1B is necessary to directly reprogram fibroblasts to induced renal tubule epithelial cells (iRECs) and, as we demonstrate, can induce ectopic pronephric tissue in Xenopus ectodermal organoids. Using these two systems, we analyzed the effect of HNF1B mutations found in patients with cystic dysplastic kidney disease. We found cross-species conserved targets of HNF1B, identified transcripts that are differentially regulated by the patient-specific mutant protein, and functionally validated novel HNF1B targets in vivo . These results highlight evolutionarily conserved transcriptional mechanisms and provide insights into the genetic circuitry of nephrogenesis.
BACKGROUND: Hepatocyte nuclear factor-1 β (HNF1B) is an essential transcription factor during embryogenesis. Mutations in HNF1B are the most common monogenic causes of congenital cystic dysplastic renal malformations. The direct functional consequences of mutations in HNF1B on its transcriptional activity are unknown.
METHODS: Direct reprogramming of mouse fibroblasts to induced renal tubular epithelial cells was conducted both with wild-type HNF1B and with patient mutations. HNF1B was expressed in Xenopus ectodermal explants. Transcriptomic analysis by bulk RNA-Seq identified conserved targets with differentially regulated expression by the wild-type or R295C mutant. CRISPR/Cas9 genome editing in Xenopus embryos evaluated transcriptional targets in vivo .
RESULTS: HNF1B is essential for reprogramming mouse fibroblasts to induced renal tubular epithelial cells and induces development of ectopic renal organoids from pluripotent Xenopus cells. The mutation R295C retains reprogramming and inductive capacity but alters the expression of specific sets of downstream target genes instead of diminishing overall transcriptional activity of HNF1B. Surprisingly, targets associated with polycystic kidney disease were less affected than genes affected in congenital renal anomalies. Cross-species-conserved transcriptional targets were dysregulated in hnf1b CRISPR-depleted Xenopus embryos, confirming their dependence on hnf1b .
CONCLUSIONS: HNF1B activates an evolutionarily conserved program of target genes that disease-causing mutations selectively disrupt. These findings provide insights into the renal transcriptional network that controls nephrogenesis.
Figure 1: Renal tissue induction in Xenopus ectodermal explants and mouse embryonic fibroblasts using transcription factors. (A) Schematics of the induction of pronephric tissue from Xenopus animal caps. Induction of pronephric tissue from animal caps treated with retinoic acid (RA) and activin A (ACT) has been previously shown by Moriya et al. (B) Results of induction efficiency using 13 renal organogenesis transcription factors and RA+ACT treatment compared with untreated (uninj.). Explants with a positive signal for the proximal tubule marker (3G8) are illustrated in the images underneath. (C) In situ stainings of distal (slc12a1/Nkcc2) and proximal (slc5a1/Sglt1) renal markers in explants induced by expression of Hnf1a, Hnf1b, Sall1, or all three factors combined compared with RA+ACT and uninjected. (D) Patient-specific mutations and the postnatal renal phenotypes illustrated on the HNF1B gene/protein schematic. (E) Schematics of the reprogramming workflow. Mouse embryonic fibroblasts were directly reprogrammed to induced renal tubular epithelial cells (iRECs) using four transcription factors (TFs)—Emx2, Hnf4a, Pax8, and HNF1B. To investigate HNF1B and its clinically relevant mutations, reprogramming with mutated HNF1B was conducted. (F) Reprogramming efficiency with patient-specific mutations. (G) Induction of pronephric tissue from animal caps with HNF1B WT and HNF1B R295C mRNA injections. (H) Proximal (3G8), distal, and connecting tubule marker (4A6) stainings in explants and a Xenopus tadpole of the same age. Positive stainings are highlighted with an arrow. Mean percentage of explants showing staining for renal markers in uninjected (n=99), injected with HNF1B WT (n=61), and explant injected with HNF1B R295C (n=91). Error bars, SEM; asterisks indicate significant differences to the positive control (uninj (B), 0TF (F) and uninjected (H)) as assessed by pairwise t-tests with corrections for multiple testing (Bonferroni), biological replicates n = 3–5 (B), n=3 (F), n=7 (H), ****P<0.0001, ***P<0.001, **P<0.01, *P<0.05. All scale bars are 200 µm.
Figure 2: Profiles of cells reprogrammed with HNF1B WT and HNF1B R295C. (A) Immunofluorescence stainings of HNF1B WT and R295C iRECs grown in 3D Matrigel for epithelial markers. Merged images consist of iRECs membrane GFP (green), target protein (red), and Hoechst (blue) signals. All scale bars are 50 µm. (B) Heatmap (left) of all the genes with significantly changed expression in at least one of the comparisons (MEFs versus 3TF+HNF1B WT, MEFs versus 3TF+HNF1B R295C, 3TF+HNF1B WT versus 3TF+HNF1B R295C) in mouse fibroblast reprogramming (N=12,742), and heatmap (right) of all the genes significant in at least one of the comparisons (uninjected versus HNF1B WT, uninjected versus HNF1B R295C and HNF1B WT versus HNF1B R295C) in explant inductions (N=16,595). (C) Venn diagrams of significantly differentially expressed (DE) up- and downregulated genes between comparisons in mouse reprogramming (blue) and explant induction (gray). (D) GO over-representation test on DE genes (absolute log2 fold change >1.5) from reprogramming experiments comparing 3TF+HNF1B WT iRECs with MEFs (top) and HNF1B WT pronephric tissue with uninjected animal caps (bottom). GO terms related to renal organogenesis are marked in bold. The same analysis for the R295C mutation condition is showed in Supplemental Figure 2A.
Figure 3: Transcriptional alterations due to HNF1B patient-specific mutation R295C. (A) Clustering DE genes according to expression patterns. Schematics represent the eight expected expression patterns while line graphs show the expression changes in the mouse transcriptome data. (B) Pie chart of the distribution of the genes in all clusters (4201 genes in total). (C) Heatmaps of top 20 most variable (variance to the mean of the cluster) genes within each cluster. (D) Comparing clusters on the GO biological process level and visualizing top three processes per cluster (full plot in Supplemental Figure 3A). (E) Percentages of genes per cluster with 1, 5, or 10-kilo base pair (K) distance from the calculated transcription starting site (data from ChIP-Seq atlas78).
Figure 4: Changes in transcription factors and genes related to kidney disease. (A) Expression patterns of transcription factors involved in kidney disease. Line graphs represent mean expression changes in three replicates along the three conditions; the plots were divided to capture the significant changes between HNF1B WT and R295C conditions. (B) Expression profile of disease-associated genes in fibroblasts and reprogrammed tissue. A significant difference in the expression levels comparing 3TF+HNF1B WT versus MEFs are represented in bold; asterisk (*) indicates a significant difference between 3TF+HNF1B R295C versus 3TF+HNF1B WT.
Figure 5: Species overlap analysis. (A) Overlap of the DE genes in mouse reprogramming and Xenopus renal tissue induction experiments. Blue ellipses indicate reprogramming from fibroblasts (MEF) with 3TF+HNF1B WT or 3TF+HNF1B R295C and the DE comparisons; gray ellipses indicate induction from animal cap (uninjected) with HNF1B WT or HNF1B R295C and the DE comparisons. (B) Heatmap of overlapping DE genes (N=117) in both species, separated on the basis of kidney-specific tissue expression in the top part (N=96). (C) Validated target genes with their function and tissue expression profile in human tissues (expression data from GTEx77).
Figure 6: In vivo analysis of target genes. (A) MesoSPIM images of the uninjected (left), control slc42a5 knockout (KO) (middle), and hnf1b KO (right) Xenopus tropicalis tadpoles and the kidney area for right (R) uninjected and left (L) manipulated sides. (B) Rescue of the hnf1b KO using WT and R295C HNF1B mRNA compared with control slc45a2 KO and uninjected conditions. Log2 changes of the kidney area from injected to uninjected sides are plotted for three different experiments (dot shape) and pairwise multiple comparisons (ANOVA) show the significance between uninjected, control injections (slc45a2 gRNA) and targeted injections. (C) In situ hybridization combined with kidney-specific antibody (lectin) stainings of six target genes in WT (left) and CRISPR/Cas9 KO (right) Xenopus embryos. (D) In situ hybridization to evaluate sord mRNA expression in CRISPR/Cas9 experiments with a gRNA (red) targeting the sord promoter at the hnf1b binding motive (light gray). Intensity of the staining was scored as weaker or stronger on the injected side or equal on both sides. Significance was calculated with the chi-square test. Scale bar is 100 μm.
Figure 7: In vitro analysis of target genes. (A) Immunofluorescence staining of HNF1B WT (left) and R295C (right) iRECs for six proteins encoded by Hnf1b-regulated genes. Merged images contain eGFP signals from the iRECs (green), antibody staining, (magenta) and nuclear stain with Hoechst (cyan). Images for all three replicates together with secondary antibodies are presented in Supplemental Figure 6A. (B) Immunofluorescence of six proteins encoded by Hnf1b-regulated genes in mouse kidney cryosections. Merged images contain autofluorescence (autofl.) of the kidney tubules (green), antibody staining (magenta), and nuclear stain with DAPI (cyan). All scale bars are 100 μm.
Supplementary Figure 1 related to Figure 1 and 2. Inducing renal tissue with HNF1B patient-specific mutation
R295C. A. Microtome sections of in situ hybridisation for slc5a1 of animal caps treated with retinoic acid and actin
(RA+ACT), extracted from tadpoles co-injected with mRNA of hnf1a, hnf1b and sall1 transcription factors (3TF) or
injected with hnf1b mRNA. Sections were stained with Eosin and DAPI. All scale bars are 50µm. B. Western blot
analysis of eight HNF1B mutations expressed in 293T cells (top/left). For the Xenopus animal cap induction
experiments HNF1B WT or R295C proteins were detected (bottom/right). C. DNA binding assay of HNF1B and
HNF1B R295C. The lower band indicates free, unbound DNA. The arrow indicates DNA bound to HNF1B proteins.
Proteins were purified from transiently transfected HEK293 cells by immunoprecipitation. DNA sequence of the
HNF1B binding motif was labelled with Cy7 which was used for detection. D. Protein stability of HNF1B WT and
HNF1B R259C mutant. Changes in protein expression of FLAG-tagged WT and mutant HNF1B upon
Cycloheximide (300μg/ul) treatment for indicated lengths of time (3-32h). E. Quantified protein expression of HNF1B
WT and HNF1B R295C normalized on Tubulin expression (±SD of minimum n = 3 independent experiments). F.
Verifying Cycloheximide activity by analyzing protein expression after 32h of treatment.
Supplementary Figure 2 related to Figure 2. RNA sequencing of the induced renal tissue. A. First two
components from principal component analysis in mouse (left) and Xenopus (right) experiments. Original tissue is
marked with the round shape and reprogrammed tissue with a triangle. Colors indicate the sample type - original
tissue (green), reprogramming with HNF1B WT (blue) and reprogramming with HNF1B R295C (red). B. Gene
Ontology (GO) over-representation test on differentially expressed genes (absolute log2 fold change >1.5) from the
reprogramming process comparing 3TF+ HNF1B R295C iRECs (left) and HNF1B R295C pronephric tissue (right)
with original material. GO terms related to renal organogenesis are marked in bold. C. Mfuzz soft clustering results
from the mouse experiments. Each line represents the expression change between original tissue, 3TF+HNF1B
WT and 3TF+HNF1B R295C conditions. Yellow colors indicate low and red high membership values (the similarity
of vectors to each other).
Supplementary Figure 3 related to Figure 3. Transcriptional alterations due to HNF1B patient-specific mutation
R295C. A. Full plot of comparing clusters on biological processes, cellular component and molecular function GO
levels. B. Distribution of expression changes between 3TF + HNF1B R295C vs 3TF + HNF1B WT in enhanced
genes in collecting duct, distal tubular and proximal tubular cells using the single-cell datasets from the Human
Protein atlas. Histograms show the genes count (left y-axis) per log2 fold change (FC) and density plot (purple)
illustrating distribution of the plot using kernel smoothing (right y-axis). C. The percentage of genes up- (red) or
downregulated (blue) in the different renal cell types.
Supplementary Figure 4 related to Figure 4. Transcriptional stability of the iRECs and genes essential for
kidney development in Xenopus organoids. A. Tubular differentiation markers Cdh6, Cdh16, Slc17a1 and Lrp2
in HNF1B WT and HNF1B R295C iRECs of later passages (passage 22) expression levels. Mean and standard
deviation of n = 3 biological replicates; differences assessed by ANOVA with Tukey’s correction for multiple
comparisons, ***P < 0.001, *P < 0.05. B. Volcano plot of HNF1B WT vs animal cap tissue DEGs with highly confident
(p-value 5e-15) genes essential for kidney development (GO:0001822 and GO:0072006). Cutoff lines for
log2foldchange are -1 and 1, for p-value 0.05.
Supplementary Figure 5 related to Figure 5. In vivo analysis of hnf1b and candidate genes. A. Expression of the
36 candidate genes from the species overlap analysis in Xenopus tropicalis. In situ hybridization was conducted at
three developmental stages - neurala, tailbud and tadpole. B. Hnf1b expression in Xenopus laevis at stages 22, 33
and 38. C. In situ hybridization combined with kidney specific LE-lectin stainings of 13 target genes in wild type and
CRISPR-Cas9 hnf1b KO Xenopus tropicalis embryos.
Supplementary Figure 6 related to Figure 6 and 7. Analysis of HNF1B targets in iRECs and in vivo. A.
Immunofluorescence stainings of three biological replicates of the reprogrammed iRECs (HNF1B WT and R295C)
for six target genes. Merged images consist of iRECs cdh16-eGFP (green), target protein (magenta) and Hoechst
(cyan) signals. B. Morphological changes of direct reprogramming using HNF1B WT (up) or HNF1B R295C (down)
as seen in differential interference contrast images. C. Genomic sequence showing the hnf1b guide RNA genome
targeted site (underlined) and sequencing chromatograms from KO and uninjected embryos. The bargraph
represents the percentages of indels occurring at the hnf1b locus in the CRISPR/Cas9 KO experiments. KO and
ICE scores based on three technical and five biological replicates.
Aboudehen,
Transcription Factor Hepatocyte Nuclear Factor-1β Regulates Renal Cholesterol Metabolism.
2016,
Pubmed
Aboudehen,
Hepatocyte Nuclear Factor-1β Regulates Urinary Concentration and Response to Hypertonicity.
2017,
Pubmed
Adalat,
HNF1B mutations associate with hypomagnesemia and renal magnesium wasting.
2009,
Pubmed
Afgan,
The Galaxy platform for accessible, reproducible and collaborative biomedical analyses: 2018 update.
2018,
Pubmed
Alvelos,
A novel mutation of the HNF1B gene associated with hypoplastic glomerulocystic kidney disease and neonatal renal failure: a case report and mutation update.
2015,
Pubmed
Andrés-Jensen,
The outcome of antenatal ultrasound diagnosed anomalies of the kidney and urinary tract in a large Danish birth cohort.
2016,
Pubmed
Attanasio,
Loss of GLIS2 causes nephronophthisis in humans and mice by increased apoptosis and fibrosis.
2007,
Pubmed
Barbacci,
HNF1beta/TCF2 mutations impair transactivation potential through altered co-regulator recruitment.
2004,
Pubmed
Barbacci,
Variant hepatocyte nuclear factor 1 is required for visceral endoderm specification.
1999,
Pubmed
Bellanné-Chantelot,
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.
2005,
Pubmed
Bellanné-Chantelot,
Clinical spectrum associated with hepatocyte nuclear factor-1beta mutations.
2004,
Pubmed
Bergmann,
Educational paper: ciliopathies.
2012,
Pubmed
Bingham,
Mutations in the hepatocyte nuclear factor-1beta gene are associated with familial hypoplastic glomerulocystic kidney disease.
2001,
Pubmed
Bohn,
Distinct molecular and morphogenetic properties of mutations in the human HNF1beta gene that lead to defective kidney development.
2003,
Pubmed
,
Xenbase
Buchholz,
Hypoxia-inducible factor-1α causes renal cyst expansion through calcium-activated chloride secretion.
2014,
Pubmed
Cereghini,
Liver-enriched transcription factors and hepatocyte differentiation.
1996,
Pubmed
Ceska,
The X-ray structure of an atypical homeodomain present in the rat liver transcription factor LFB1/HNF1 and implications for DNA binding.
1993,
Pubmed
Chan,
Mechanism of Fibrosis in HNF1B-Related Autosomal Dominant Tubulointerstitial Kidney Disease.
2018,
Pubmed
Chen,
VennDiagram: a package for the generation of highly-customizable Venn and Euler diagrams in R.
2011,
Pubmed
Clissold,
HNF1B-associated renal and extra-renal disease-an expanding clinical spectrum.
2015,
Pubmed
Clissold,
Genome-wide methylomic analysis in individuals with HNF1B intragenic mutation and 17q12 microdeletion.
2018,
Pubmed
Decramer,
Anomalies of the TCF2 gene are the main cause of fetal bilateral hyperechogenic kidneys.
2007,
Pubmed
Desgrange,
HNF1B controls epithelial organization and cell polarity during ureteric bud branching and collecting duct morphogenesis.
2017,
Pubmed
Dobin,
STAR: ultrafast universal RNA-seq aligner.
2013,
Pubmed
Drews,
The nephrogenic potential of the transcription factors osr1, osr2, hnf1b, lhx1 and pax8 assessed in Xenopus animal caps.
2011,
Pubmed
,
Xenbase
Drost,
Biomartr: genomic data retrieval with R.
2017,
Pubmed
Eckardt,
Autosomal dominant tubulointerstitial kidney disease: diagnosis, classification, and management--A KDIGO consensus report.
2015,
Pubmed
Faguer,
Hnf-1β transcription factor is an early hif-1α-independent marker of epithelial hypoxia and controls renal repair.
2013,
Pubmed
Faguer,
Diagnosis, management, and prognosis of HNF1B nephropathy in adulthood.
2011,
Pubmed
Falk,
U-Net: deep learning for cell counting, detection, and morphometry.
2019,
Pubmed
Ferrè,
HNF-1B specifically regulates the transcription of the γa-subunit of the Na+/K+-ATPase.
2011,
Pubmed
Ferrè,
Mutations in PCBD1 cause hypomagnesemia and renal magnesium wasting.
2014,
Pubmed
Ferrè,
New insights into the role of HNF-1β in kidney (patho)physiology.
2019,
Pubmed
Gentleman,
Bioconductor: open software development for computational biology and bioinformatics.
2004,
Pubmed
Getwan,
Toolbox in a tadpole: Xenopus for kidney research.
2017,
Pubmed
,
Xenbase
Gresh,
A transcriptional network in polycystic kidney disease.
2004,
Pubmed
GTEx Consortium,
The Genotype-Tissue Expression (GTEx) project.
2013,
Pubmed
Gu,
Complex heatmaps reveal patterns and correlations in multidimensional genomic data.
2016,
Pubmed
Heidet,
Spectrum of HNF1B mutations in a large cohort of patients who harbor renal diseases.
2010,
Pubmed
Heliot,
HNF1B controls proximal-intermediate nephron segment identity in vertebrates by regulating Notch signalling components and Irx1/2.
2013,
Pubmed
,
Xenbase
Hiesberger,
Mutation of hepatocyte nuclear factor-1beta inhibits Pkhd1 gene expression and produces renal cysts in mice.
2004,
Pubmed
Hiesberger,
Role of the hepatocyte nuclear factor-1beta (HNF-1beta) C-terminal domain in Pkhd1 (ARPKD) gene transcription and renal cystogenesis.
2005,
Pubmed
Hildebrandt,
Ciliopathies.
2011,
Pubmed
Hildebrandt,
Nephronophthisis: disease mechanisms of a ciliopathy.
2009,
Pubmed
Hoff,
ANKS6 is a central component of a nephronophthisis module linking NEK8 to INVS and NPHP3.
2013,
Pubmed
,
Xenbase
Horikawa,
Mutation in hepatocyte nuclear factor-1 beta gene (TCF2) associated with MODY.
1997,
Pubmed
Huber,
Orchestrating high-throughput genomic analysis with Bioconductor.
2015,
Pubmed
Kaminski,
Direct reprogramming of fibroblasts into renal tubular epithelial cells by defined transcription factors.
2016,
Pubmed
,
Xenbase
Karczewski,
The mutational constraint spectrum quantified from variation in 141,456 humans.
2020,
Pubmed
Karimi,
Xenbase: a genomic, epigenomic and transcriptomic model organism database.
2018,
Pubmed
,
Xenbase
Kompatscher,
Loss of transcriptional activation of the potassium channel Kir5.1 by HNF1β drives autosomal dominant tubulointerstitial kidney disease.
2017,
Pubmed
Kumar,
Mfuzz: a software package for soft clustering of microarray data.
2007,
Pubmed
Kumaran,
Identification and classification of epithelial cells in nephron segments by actin cytoskeleton patterns.
2020,
Pubmed
Lamb,
Neural induction by the secreted polypeptide noggin.
1993,
Pubmed
,
Xenbase
Laurichesse Delmas,
Congenital unilateral renal agenesis: Prevalence, prenatal diagnosis, associated anomalies. Data from two birth-defect registries.
2017,
Pubmed
Lea,
A new epitope-tagged Pkhd1 allele sheds light on fibrocystin signaling.
2017,
Pubmed
Liao,
featureCounts: an efficient general purpose program for assigning sequence reads to genomic features.
2014,
Pubmed
Lienkamp,
Inversin relays Frizzled-8 signals to promote proximal pronephros development.
2010,
Pubmed
,
Xenbase
Lienkamp,
Vertebrate kidney tubules elongate using a planar cell polarity-dependent, rosette-based mechanism of convergent extension.
2012,
Pubmed
,
Xenbase
Lienkamp,
Using Xenopus to study genetic kidney diseases.
2016,
Pubmed
,
Xenbase
Limaye,
Expression of specific hepatocyte and cholangiocyte transcription factors in human liver disease and embryonic development.
2008,
Pubmed
Lokmane,
vHNF1 functions in distinct regulatory circuits to control ureteric bud branching and early nephrogenesis.
2010,
Pubmed
Love,
Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2.
2014,
Pubmed
Lu,
Structural basis of disease-causing mutations in hepatocyte nuclear factor 1beta.
2007,
Pubmed
Lu,
Mutations in DZIP1L, which encodes a ciliary-transition-zone protein, cause autosomal recessive polycystic kidney disease.
2017,
Pubmed
Lu,
HNF1B inhibits cell proliferation via repression of SMAD6 expression in prostate cancer.
2020,
Pubmed
Marchesin,
Molecular Basis for Autosomal-Dominant Renal Fanconi Syndrome Caused by HNF4A.
2019,
Pubmed
Miner,
Defective glomerulogenesis in the absence of laminin alpha5 demonstrates a developmental role for the kidney glomerular basement membrane.
2000,
Pubmed
Mochizuki,
PKD2, a gene for polycystic kidney disease that encodes an integral membrane protein.
1996,
Pubmed
Moriya,
Induction of Pronephric Tubules by Activin and Retinoic Acid in Presumptive Ectoderm of Xenopus laevis: (RA/kidney/mesoderm induction/Xenopus laevis).
1993,
Pubmed
,
Xenbase
Naert,
Deep learning is widely applicable to phenotyping embryonic development and disease.
2021,
Pubmed
,
Xenbase
Nakayama,
Simple and efficient CRISPR/Cas9-mediated targeted mutagenesis in Xenopus tropicalis.
2013,
Pubmed
,
Xenbase
Nakayama,
HNF1B alterations associated with congenital anomalies of the kidney and urinary tract.
2010,
Pubmed
Niborski,
Hnf1b haploinsufficiency differentially affects developmental target genes in a new renal cysts and diabetes mouse model.
2021,
Pubmed
Nigam,
Impact of next generation sequencing on our understanding of CAKUT.
2019,
Pubmed
NULL,
The polycystic kidney disease 1 gene encodes a 14 kb transcript and lies within a duplicated region on chromosome 16. The European Polycystic Kidney Disease Consortium.
1994,
Pubmed
Oki,
ChIP-Atlas: a data-mining suite powered by full integration of public ChIP-seq data.
2018,
Pubmed
Pollard,
Targeted inactivation of fh1 causes proliferative renal cyst development and activation of the hypoxia pathway.
2007,
Pubmed
Pontén,
The Human Protein Atlas--a tool for pathology.
2008,
Pubmed
Raciti,
Organization of the pronephric kidney revealed by large-scale gene expression mapping.
2008,
Pubmed
,
Xenbase
Robinson,
A Hypoxia-Inducible HIF1-GAL3ST1-Sulfatide Axis Enhances ccRCC Immune Evasion via Increased Tumor Cell-Platelet Binding.
2019,
Pubmed
Roelandt,
HNF1B deficiency causes ciliary defects in human cholangiocytes.
2012,
Pubmed
Sander,
Transcriptional profiling of the zebrafish proximal tubule.
2019,
Pubmed
Schindelin,
Fiji: an open-source platform for biological-image analysis.
2012,
Pubmed
Schmittgen,
Analyzing real-time PCR data by the comparative C(T) method.
2008,
Pubmed
Seikaly,
Chronic renal insufficiency in children: the 2001 Annual Report of the NAPRTCS.
2003,
Pubmed
Shen,
A map of the cis-regulatory sequences in the mouse genome.
2012,
Pubmed
Stanley,
Causal Genetic Variants in Stillbirth.
2020,
Pubmed
Steenhard,
Transgenic expression of human LAMA5 suppresses murine Lama5 mRNA and laminin α5 protein deposition.
2011,
Pubmed
Stettner,
Sulfatides are required for renal adaptation to chronic metabolic acidosis.
2013,
Pubmed
Strazzabosco,
Development of the bile ducts: essentials for the clinical hepatologist.
2012,
Pubmed
Tanaka,
Hypoxia in renal disease with proteinuria and/or glomerular hypertension.
2004,
Pubmed
Teo,
Early Developmental Perturbations in a Human Stem Cell Model of MODY5/HNF1B Pancreatic Hypoplasia.
2016,
Pubmed
Thomas,
HNF1B and PAX2 mutations are a common cause of renal hypodysplasia in the CKiD cohort.
2011,
Pubmed
Tran,
Xenopus Bicaudal-C is required for the differentiation of the amphibian pronephros.
2007,
Pubmed
,
Xenbase
Ulinski,
Renal phenotypes related to hepatocyte nuclear factor-1beta (TCF2) mutations in a pediatric cohort.
2006,
Pubmed
van der Ven,
Whole-Exome Sequencing Identifies Causative Mutations in Families with Congenital Anomalies of the Kidney and Urinary Tract.
2018,
Pubmed
Voigt,
The mesoSPIM initiative: open-source light-sheet microscopes for imaging cleared tissue.
2019,
Pubmed
Wang,
Selective deletion of the Hnf1beta (MODY5) gene in beta-cells leads to altered gene expression and defective insulin release.
2004,
Pubmed
Wang,
HNF1B-mediated repression of SLUG is suppressed by EZH2 in aggressive prostate cancer.
2020,
Pubmed
Weber,
Prevalence of mutations in renal developmental genes in children with renal hypodysplasia: results of the ESCAPE study.
2006,
Pubmed
Yoder,
The polycystic kidney disease proteins, polycystin-1, polycystin-2, polaris, and cystin, are co-localized in renal cilia.
2002,
Pubmed
Yu,
clusterProfiler: an R package for comparing biological themes among gene clusters.
2012,
Pubmed
Zaucke,
Uromodulin is expressed in renal primary cilia and UMOD mutations result in decreased ciliary uromodulin expression.
2010,
Pubmed
Zhou,
Proximo-distal specialization of epithelial transport processes within the Xenopus pronephric kidney tubules.
2004,
Pubmed
,
Xenbase