Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 599
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-35020897

ABSTRACT

The patterned array of basal, intermediate and superficial cells in the urothelium of the mature ureter arises from uncommitted epithelial progenitors of the distal ureteric bud. Urothelial development requires signaling input from surrounding mesenchymal cells, which, in turn, depend on cues from the epithelial primordium to form a layered fibro-muscular wall. Here, we have identified FGFR2 as a crucial component in this reciprocal signaling crosstalk in the murine ureter. Loss of Fgfr2 in the ureteric epithelium led to reduced proliferation, stratification, intermediate and basal cell differentiation in this tissue, and affected cell survival and smooth muscle cell differentiation in the surrounding mesenchyme. Loss of Fgfr2 impacted negatively on epithelial expression of Shh and its mesenchymal effector gene Bmp4. Activation of SHH or BMP4 signaling largely rescued the cellular defects of mutant ureters in explant cultures. Conversely, inhibition of SHH or BMP signaling in wild-type ureters recapitulated the mutant phenotype in a dose-dependent manner. Our study suggests that FGF signals from the mesenchyme enhance, via epithelial FGFR2, the SHH-BMP4 signaling axis to drive urothelial and mesenchymal development in the early ureter.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Hedgehog Proteins/metabolism , Organogenesis , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction , Ureter/metabolism , Animals , Mesoderm/cytology , Mesoderm/metabolism , Mice , Receptor, Fibroblast Growth Factor, Type 2/genetics , Ureter/embryology , Urothelium/cytology , Urothelium/metabolism
2.
Pediatr Res ; 95(7): 1754-1757, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38287105

ABSTRACT

BACKGROUND: The prorenin receptor (PRR) plays a critical role in ureteric bud (UB) branching morphogenesis. DOT1 Like (DOT1L), a histone methyltransferase specific for Histone 3 lysine 79 (H3K79), is important for differentiation of the UB-derived renal collecting duct cells. In this study, we tested whether DOT1L/H3 dimethyl K79 (H3m2K79) are regulated by PRR deletion in the UB and UB-derived collecting ducts in the embryonic mouse kidneys. METHODS: Mutant Hoxb7Cre+/PRRflox/flox (PRRUB-/-) and control PRRUB+/+, mice were studied on embryonic (E) day E17.5. DOT1L mRNA and protein expression in the kidney was examined by real-time qRT-PCR and immunohistochemistry, respectively. H3m2K79 protein expression was determined by immunohistochemistry and Western blot analysis. RESULTS: DOT1L mRNA levels were decreased in mutant compared to control mice (0.68 ± 0.06 vs. 1.0 ± 0.01, p < 0.01). DOT1L and H3m2K79 immunostaining was reduced in the mutant vs. control kidneys (Dot1: 0.62 ± 0.03 vs. 1.0 ± 0.01, p < 0.05; H3m2K79: 0.64 ± 0.04 vs.1.1 ± 0.01. p < 0.05.). Western blot analysis revealed decreased H3m2K79 protein levels in mutant compared to control kidneys (1.0 ± 0.06 vs. 1.5 ± 0.02, p < 0.05). CONCLUSION: Targeted deletion of the PRR in the UB and UB-derived collecting ducts results in reduced DOT1L gene/protein and H3m2K79 protein expression in the embryonic mouse metanephroi in vivo. IMPACT: The role of histone methylation in mediating the effect of the prorenin receptor on the ureteric bud branching (UB) morphogenesis and urine acidification during kidney development is unknown. We demonstrate that histone H3 lysine (K) 79 dimethylation by methyltransferase Dot1 is reduced in the embryonic kidney of mice that lack the prorenin receptor in the UB lineage.


Subject(s)
Histone-Lysine N-Methyltransferase , Histones , Prorenin Receptor , Receptors, Cell Surface , Ureter , Animals , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Mice , Histones/metabolism , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , Ureter/embryology , Ureter/metabolism , Signal Transduction , Mice, Knockout , Gene Deletion , Methylation , Kidney/metabolism , Kidney/embryology , RNA, Messenger/metabolism , RNA, Messenger/genetics , Gene Expression Regulation, Developmental , Kidney Tubules, Collecting/metabolism , Kidney Tubules, Collecting/embryology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Embryonic Structures
3.
Biochem J ; 479(1): 91-109, 2022 01 14.
Article in English | MEDLINE | ID: mdl-34935912

ABSTRACT

The TBX18 transcription factor regulates patterning and differentiation programs in the primordia of many organs yet the molecular complexes in which TBX18 resides to exert its crucial transcriptional function in these embryonic contexts have remained elusive. Here, we used 293 and A549 cells as an accessible cell source to search for endogenous protein interaction partners of TBX18 by an unbiased proteomic approach. We tagged endogenous TBX18 by CRISPR/Cas9 targeted genome editing with a triple FLAG peptide, and identified by anti-FLAG affinity purification and subsequent LC-MS analysis the ZMYM2 protein to be statistically enriched together with TBX18 in both 293 and A549 nuclear extracts. Using a variety of assays, we confirmed the binding of TBX18 to ZMYM2, a component of the CoREST transcriptional corepressor complex. Tbx18 is coexpressed with Zmym2 in the mesenchymal compartment of the developing ureter of the mouse, and mutations in TBX18 and in ZMYM2 were recently linked to congenital anomalies in the kidney and urinary tract (CAKUT) in line with a possible in vivo relevance of TBX18-ZMYM2 protein interaction in ureter development.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Proteomics/methods , Signal Transduction/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , A549 Cells , Animals , DNA-Binding Proteins/genetics , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Humans , Mice , Mutation , Pregnancy , Protein Binding , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Transfection , Ureter/embryology , Ureter/metabolism , Urogenital Abnormalities/genetics , Urogenital Abnormalities/metabolism , Vesico-Ureteral Reflux/genetics , Vesico-Ureteral Reflux/metabolism
4.
Kidney Int ; 102(1): 108-120, 2022 07.
Article in English | MEDLINE | ID: mdl-35341793

ABSTRACT

Oxidative metabolism in mitochondria regulates cellular differentiation and gene expression through intermediary metabolites and reactive oxygen species. Its role in kidney development and pathogenesis is not completely understood. Here we inactivated ubiquinone-binding protein QPC, a subunit of mitochondrial complex III, in two types of kidney progenitor cells to investigate the role of mitochondrial electron transport in kidney homeostasis. Inactivation of QPC in sine oculis-related homeobox 2 (SIX2)-expressing cap mesenchyme progenitors, which give rise to podocytes and all nephron segments except collecting ducts, resulted in perinatal death from severe kidney dysplasia. This was characterized by decreased proliferation of SIX2 progenitors and their failure to differentiate into kidney epithelium. QPC inactivation in cap mesenchyme progenitors induced activating transcription factor 4-mediated nutritional stress responses and was associated with a reduction in kidney tricarboxylic acid cycle metabolites and amino acid levels, which negatively impacted purine and pyrimidine synthesis. In contrast, QPC inactivation in ureteric tree epithelial cells, which give rise to the kidney collecting system, did not inhibit ureteric differentiation, and resulted in the development of functional kidneys that were smaller in size. Thus, our data demonstrate that mitochondrial oxidative metabolism is critical for the formation of cap mesenchyme-derived nephron segments but dispensable for formation of the kidney collecting system. Hence, our studies reveal compartment-specific needs for metabolic reprogramming during kidney development.


Subject(s)
Electron Transport Complex III , Kidney , Nephrons , Organogenesis , Podocytes , Amino Acids/deficiency , Cell Differentiation , Electron Transport Complex III/metabolism , Female , Humans , Kidney/embryology , Kidney/metabolism , Mesoderm/metabolism , Nephrons/metabolism , Organogenesis/genetics , Podocytes/metabolism , Pregnancy , Ureter/embryology
5.
Biochem Biophys Res Commun ; 589: 173-179, 2022 01 22.
Article in English | MEDLINE | ID: mdl-34922199

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) are a family of often-concurrent diseases with various anatomical spectra. Null-mutant Gen1 mice frequently develop multiple urinary phenotypes, most commonly duplex kidneys, and are ideal subjects for research on ectopic budding in CAKUT development. The upper and lower kidney poles of the Gen1PB/PB mouse were examined by histology, immunofluorescence, and immunohistochemistry. The newborn Gen1PB/PB mouse lower poles were significantly more hypoplastic than the corresponding upper poles, with significantly fewer glomeruli. On embryonic day 14.5, immediately before first urine formation, the upper pole kidney was already larger than the lower pole kidney. In vivo and in vitro, embryonic kidney upper poles had more ureteric buds than lower poles. Gen1PB/PB embryos exhibited ectopic ureteric buds, usually near the original budding site, occasionally far away, or, rarely, derived from the primary budding site. Therefore, ectopia of the ureteric buds is the core of CAKUT formation. Further studies will be needed to investigate the regulatory roles of these genes in initial ureteric budding and subsequent ontogenesis during metanephros development.


Subject(s)
Holliday Junction Resolvases/metabolism , Kidney/abnormalities , Kidney/embryology , Animals , Animals, Newborn , Biomarkers/metabolism , Cell Differentiation , Embryo, Mammalian/pathology , Mice , Ureter/abnormalities , Ureter/embryology
6.
Development ; 146(12)2019 06 12.
Article in English | MEDLINE | ID: mdl-31118232

ABSTRACT

Recent advances in the generation of kidney organoids and the culture of primary nephron progenitors from mouse and human have been based on knowledge of the molecular basis of kidney development in mice. Although gene expression during kidney development has been intensely investigated, single cell profiling provides new opportunities to further subsect component cell types and the signalling networks at play. Here, we describe the generation and analysis of 6732 single cell transcriptomes from the fetal mouse kidney [embryonic day (E)18.5] and 7853 sorted nephron progenitor cells (E14.5). These datasets provide improved resolution of cell types and specific markers, including subdivision of the renal stroma and heterogeneity within the nephron progenitor population. Ligand-receptor interaction and pathway analysis reveals novel crosstalk between cellular compartments and associates new pathways with differentiation of nephron and ureteric epithelium cell types. We identify transcriptional congruence between the distal nephron and ureteric epithelium, showing that most markers previously used to identify ureteric epithelium are not specific. Together, this work improves our understanding of metanephric kidney development and provides a template to guide the regeneration of renal tissue.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Kidney/embryology , Receptor Cross-Talk , Single-Cell Analysis/methods , Algorithms , Animals , Cell Differentiation , Cell Lineage , Epithelium/embryology , Kidney/cytology , Ligands , Mice , Mice, Inbred C57BL , Nephrons/embryology , Organogenesis , Signal Transduction , Stem Cells/cytology , Transcriptome , Ureter/embryology
7.
Dev Biol ; 464(2): 103-110, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32562756

ABSTRACT

Congenital anomalies of the urinary tract are a significant cause of morbidity in infancy, and many congenital anomalies are linked to ureter development; however, the mechanism by which congenital anomalies control ureter development remains unknown. The loss of Robo2 can cause ureter defects and vesicoureteral reflux. However, how Robo2 impacts ureter development is unclear. We found that ROBO2 is expressed in the common nephric duct (CND) and primitive bladder, and impacts CND migration and fusion with the primitive bladder via its novel binding partner retinaldehyde dehydrogenase-2 (RALDH2). Delayed apoptosis that is due to the failure of CND fusion with the primitive bladder in the Robo2-/-embryo results in an abnormal ureter connection to the CND, which is required for ureter development. We define a novel pathway in which the CND is remodeled by ROBO2 and retinoic acid rescued the ureter anomalies in the Robo2-/-embryo. These findings may be relevant to diverse disease conditions that are associated with altered signaling in the primitive bladder.


Subject(s)
Aldehyde Oxidoreductases/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Ureter/embryology , Urinary Bladder/embryology , Aldehyde Oxidoreductases/genetics , Animals , Mice , Mice, Knockout , Receptors, Immunologic/genetics , Ureter/cytology , Urinary Bladder/cytology
8.
Dev Biol ; 465(1): 23-30, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32645357

ABSTRACT

Congenital anomalies of external genitalia affect approximately 1 in 125 live male births. Development of the genital tubercle, the precursor of the penis and clitoris, is regulated by the urethral plate epithelium, an endodermal signaling center. Signaling activity of the urethral plate is mediated by Sonic hedgehog (SHH), which coordinates outgrowth and patterning of the genital tubercle by controlling cell cycle kinetics and expression of downstream genes. The mechanisms that govern Shh transcription in urethral plate cells are largely unknown. Here we show that deletion of Foxa1 and Foxa2 results in persistent cloaca, an incomplete separation of urinary, genital, and anorectal tracts, and severe hypospadias, a failure of urethral tubulogenesis. Loss of Foxa2 and only one copy of Foxa1 results in urethral fistula, an additional opening of the penile urethra. Foxa1/a2 participate in an autoregulatory feedback loop with Shh, in which FOXA1 and FOXA2 positively regulate transcription of Shh in the urethra, and SHH feeds back to negatively regulate Foxa1 and Foxa2 expression. These findings reveal novel roles for Foxa genes in development of the urethral tube and in division of the embryonic cloaca.


Subject(s)
Cloaca/embryology , Embryo, Mammalian/embryology , Hedgehog Proteins/metabolism , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-beta/metabolism , Ureter/embryology , Animals , Hedgehog Proteins/genetics , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-beta/genetics , Mice , Mice, Transgenic
9.
Dev Biol ; 458(2): 164-176, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31734175

ABSTRACT

The ureteric epithelial progenitor (UEP) population within the embryonic kidney generates the arborized epithelial network of the kidney's collecting system and plays a critical role in the expansion and induction of the surrounding nephron progenitor pool. Adamts18 shows UEP- restricted expression in the kidney and progenitor tip-restricted expression in several other organs undergoing branching epithelial growth. Adamts18 is encoded by 23 exons. Genetic removal of genomic sequence spanning exons 1 to 3 led to a specific loss of Adamts18 expression in UEPs, suggesting this region may encode a UEP-specific enhancer. Intron 2 (3 â€‹kb) was shown to have enhancer activity driving expression of the doxycycline inducible tet-on transcriptional regulator (rtTA) in an Adamts18en-rtTA transgenic mouse strain. Crossing Adamts18en-rtTA mice to a doxycycline dependent GFP reporter mouse enabled the live imaging of embryonic kidney explants. This facilitated the analysis of ureteric epithelial branching events at the cellular level. Ablation of UEPs at the initiation of ureteric bud outgrowth through the doxycycline-mediated induction of Diphtheria Toxin A (DTA) generated a range of phenotypes from complete kidneys agenesis, to duplex kidneys with double ureters. The latter outcome points to the potential of regulative processes to restore UEPs. In contrast, overexpression of YAP prior to ureteric bud outgrowth led to a complete failure of kidney development. Elevating YAP levels at later stages retarded branching growth. A similar phenotype was observed with the overexpression of MYC within the branch-tip localized UEP population. These experiments showcase the utility of the Adamts18en-rtTA transgenic model to the investigation of cellular and molecular events specific to branch tip progenitors within the mammalian kidney complementing existing CRE-dependent genetic tools. Further, the illustrative examples point to areas where new insight may be gained into the regulation of UEP programs.


Subject(s)
ADAMTS Proteins/genetics , ADAMTS Proteins/metabolism , Ureter/embryology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins/metabolism , Female , Kidney/metabolism , Kidney/pathology , Male , Mammals/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Morphogenesis/genetics , Nephrons/metabolism , Organogenesis/genetics , Regulatory Sequences, Nucleic Acid/genetics , Ureter/metabolism , YAP-Signaling Proteins
10.
Development ; 145(23)2018 11 26.
Article in English | MEDLINE | ID: mdl-30478225

ABSTRACT

The organized array of smooth muscle cells (SMCs) and fibroblasts in the walls of visceral tubular organs arises by patterning and differentiation of mesenchymal progenitors surrounding the epithelial lumen. Here, we show that the TBX2 and TBX3 transcription factors have novel and required roles in regulating these processes in the murine ureter. Co-expression of TBX2 and TBX3 in the inner mesenchymal region of the developing ureter requires canonical WNT signaling. Loss of TBX2/TBX3 in this region disrupts activity of two crucial drivers of the SMC program, Foxf1 and BMP4 signaling, resulting in decreased SMC differentiation and increased extracellular matrix. Transcriptional profiling and chromatin immunoprecipitation experiments revealed that TBX2/TBX3 directly repress expression of the WNT antagonists Dkk2 and Shisa2, the BMP antagonist Bmper and the chemokine Cxcl12 These findings suggest that TBX2/TBX3 are effectors of canonical WNT signaling in the ureteric mesenchyme that promote SMC differentiation by maintaining BMP4 and WNT signaling in the inner region, while restricting CXCL12 signaling to the outer layer of fibroblast-fated mesenchyme.


Subject(s)
Body Patterning , Cell Differentiation , Mesoderm/embryology , T-Box Domain Proteins/metabolism , Ureter/embryology , Wnt Signaling Pathway , Animals , Bone Morphogenetic Protein 4/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Mice , Models, Biological , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Peristalsis , T-Box Domain Proteins/genetics , Transcriptome/genetics , Ureter/metabolism , Ureter/pathology
11.
J Pathol ; 252(3): 290-303, 2020 11.
Article in English | MEDLINE | ID: mdl-32715478

ABSTRACT

Development of the mammalian kidney is orchestrated by reciprocal interactions of stromal and nephrogenic mesenchymal cells with the ureteric bud epithelium. Previous work showed that the transcription factor Wilms tumor 1 (WT1) acts in the nephrogenic lineage to maintain precursor cells, to drive the epithelial transition of aggregating precursors into a renal vesicle and to specify and maintain the podocyte fate. However, WT1 is expressed not only in the nephrogenic lineage but also transiently in stromal progenitors in the renal cortex. Here we report that specific deletion of Wt1 in the stromal lineage using the Foxd1cre driver line results at birth in cryptorchidism and hypoplastic kidneys that harbour fewer and enlarged ureteric bud tips and display an expansion of capsular stroma into the cortical region. In vivo and ex vivo analysis at earlier stages revealed that stromal loss of Wt1 reduces stromal proliferation, and delays and alters branching morphogenesis, resulting in a variant architecture of the collecting duct tree with an increase of single at the expense of bifurcated ureteric bud tips. Molecular analysis identified a transient reduction of Aldh1a2 expression and of retinoic acid signalling activity in stromal progenitors, and of Ret in ureteric bud tips. Administration of retinoic acid partly rescued the branching defects of mutant kidneys in culture. We propose that WT1 maintains retinoic acid signalling in the cortical stroma, which, in turn, assures proper levels and dynamics of Ret expression in the ureteric bud tips, and thus normal ramification of the ureteric tree. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Cryptorchidism/embryology , Cryptorchidism/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Kidney/embryology , Ureter/embryology , WT1 Proteins/genetics , Animals , Biomarkers/metabolism , Cryptorchidism/metabolism , Kidney/abnormalities , Kidney/metabolism , Male , Mice , Organogenesis/genetics , Ureter/abnormalities , Ureter/metabolism , WT1 Proteins/metabolism
12.
Development ; 144(17): 3177-3188, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28705898

ABSTRACT

Branching morphogenesis creates arborized epithelial networks. In the mammalian kidney, an epithelial progenitor pool at ureteric branch tips (UBTs) creates the urine-transporting collecting system. Using region-specific mouse reporter strains, we performed an RNA-seq screen, identifying tip- and stalk-enriched gene sets in the developing collecting duct system. Detailed in situ hybridization studies of tip-enriched predictions identified UBT-enriched gene sets conserved between the mouse and human kidney. Comparative spatial analysis of their UBT niche expression highlighted distinct patterns of gene expression revealing novel molecular heterogeneity within the UBT progenitor population. To identify kidney-specific and shared programs of branching morphogenesis, comparative expression studies on the developing mouse lung were combined with in silico analysis of the developing mouse salivary gland. These studies highlight a shared gene set with multi-organ tip enrichment and a gene set specific to UBTs. This comprehensive analysis extends our current understanding of the ureteric branch tip niche.


Subject(s)
Organogenesis , Stem Cell Niche , Ureter/cytology , Ureter/embryology , Animals , Gene Expression Regulation, Developmental , Humans , In Situ Hybridization , Kidney/embryology , Kidney/metabolism , Mice , Organ Specificity/genetics , Organogenesis/genetics , Sequence Analysis, RNA , Stem Cell Niche/genetics
13.
Development ; 144(22): 4148-4158, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28993400

ABSTRACT

Kidney collecting system development requires integrin-dependent cell-extracellular matrix interactions. Integrins are heterodimeric transmembrane receptors consisting of α and ß subunits; crucial integrins in the kidney collecting system express the ß1 subunit. The ß1 cytoplasmic tail has two NPxY motifs that mediate functions by binding to cytoplasmic signaling and scaffolding molecules. Talins, scaffolding proteins that bind to the membrane proximal NPxY motif, are proposed to activate integrins and to link them to the actin cytoskeleton. We have defined the role of talin binding to the ß1 proximal NPxY motif in the developing kidney collecting system in mice that selectively express a Y-to-A mutation in this motif. The mice developed a hypoplastic dysplastic collecting system. Collecting duct cells expressing this mutation had moderate abnormalities in cell adhesion, migration, proliferation and growth factor-dependent signaling. In contrast, mice lacking talins in the developing ureteric bud developed kidney agenesis and collecting duct cells had severe cytoskeletal, adhesion and polarity defects. Thus, talins are essential for kidney collecting duct development through mechanisms that extend beyond those requiring binding to the ß1 integrin subunit NPxY motif.


Subject(s)
Integrin beta1/metabolism , Morphogenesis , Talin/metabolism , Ureter/cytology , Ureter/embryology , Adherens Junctions/metabolism , Amino Acid Motifs , Animals , Binding Sites , Cell Adhesion , Cell Membrane/metabolism , Cell Polarity , Gene Expression Regulation, Developmental , Integrin beta1/chemistry , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/embryology , Mice, Inbred C57BL , Mutation/genetics , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Ureter/metabolism
14.
Development ; 144(17): 3080-3094, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28760814

ABSTRACT

The formation of the proper number of nephrons requires a tightly regulated balance between renal progenitor cell self-renewal and differentiation. The molecular pathways that regulate the transition from renal progenitor to renal vesicle are not well understood. Here, we show that Sall1interacts with the nucleosome remodeling and deacetylase complex (NuRD) to inhibit premature differentiation of nephron progenitor cells. Disruption of Sall1-NuRD in vivo in knock-in mice (ΔSRM) resulted in accelerated differentiation of nephron progenitors and bilateral renal hypoplasia. Transcriptional profiling of mutant kidneys revealed a striking pattern in which genes of the glomerular and proximal tubule lineages were either unchanged or upregulated, and those in the loop of Henle and distal tubule lineages were downregulated. These global changes in gene expression were accompanied by a significant decrease in THP-, NKCC2- and AQP1-positive loop of Henle nephron segments in mutant ΔSRM kidneys. These findings highlight an important function of Sall1-NuRD interaction in the regulation of Six2-positive multipotent renal progenitor cells and formation of the loop of Henle.


Subject(s)
Loop of Henle/embryology , Loop of Henle/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Multipotent Stem Cells/cytology , Organogenesis , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Gene Ontology , Homozygote , Kidney Tubules/metabolism , Loop of Henle/abnormalities , Mice , Multipotent Stem Cells/metabolism , Mutation/genetics , Organogenesis/genetics , Protein Binding/genetics , Transcription Factors/chemistry , Ureter/embryology , Ureter/metabolism
15.
Development ; 144(24): 4704-4719, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29158444

ABSTRACT

Kidney development depends crucially on proper ureteric bud branching giving rise to the entire collecting duct system. The transcription factor HNF1B is required for the early steps of ureteric bud branching, yet the molecular and cellular events regulated by HNF1B are poorly understood. We report that specific removal of Hnf1b from the ureteric bud leads to defective cell-cell contacts and apicobasal polarity during the early branching events. High-resolution ex vivo imaging combined with a membranous fluorescent reporter strategy show decreased mutant cell rearrangements during mitosis-associated cell dispersal and severe epithelial disorganization. Molecular analysis reveals downregulation of Gdnf-Ret pathway components and suggests that HNF1B acts both upstream and downstream of Ret signaling by directly regulating Gfra1 and Etv5 Subsequently, Hnf1b deletion leads to massively mispatterned ureteric tree network, defective collecting duct differentiation and disrupted tissue architecture, which leads to cystogenesis. Consistently, mRNA-seq analysis shows that the most impacted genes encode intrinsic cell-membrane components with transporter activity. Our study uncovers a fundamental and recurring role of HNF1B in epithelial organization during early ureteric bud branching and in further patterning and differentiation of the collecting duct system in mouse.


Subject(s)
Cell Polarity/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney Tubules, Collecting/embryology , Ureter/embryology , Urogenital Abnormalities/embryology , Urogenital Abnormalities/genetics , Animals , Cell Adhesion/genetics , Cells, Cultured , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Hepatocyte Nuclear Factor 1-beta/metabolism , Mice , Mice, Knockout , Nuclear Proteins/metabolism , Organ Culture Techniques , PAX2 Transcription Factor/biosynthesis , Signal Transduction/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases
16.
Development ; 144(23): 4377-4385, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29038307

ABSTRACT

Metanephric kidney development is orchestrated by the iterative branching morphogenesis of the ureteric bud. We describe an underlying patterning associated with the ramification of this structure and show that this pattern is conserved between developing kidneys, in different parts of the organ and across developmental time. This regularity is associated with a highly reproducible branching asymmetry that is consistent with locally operative growth mechanisms. We then develop a class of tip state models to represent elaboration of the ureteric tree and describe rules for 'half-delay' branching morphogenesis that describe almost perfectly the patterning of this structure. Spatial analysis suggests that the observed asymmetry may arise from mutual suppression of bifurcation, but not extension, between the growing ureteric tips, and demonstrates that disruption of patterning occurs in mouse mutants in which the distribution of tips on the surface of the kidney is altered. These findings demonstrate that kidney development occurs by way of a highly conserved reiterative pattern of asymmetric bifurcation that is governed by intrinsic and locally operative mechanisms.


Subject(s)
Kidney/embryology , Morphogenesis/physiology , Ureter/embryology , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/physiology , Animals , Body Patterning/genetics , Body Patterning/physiology , Bone Morphogenetic Protein 7/deficiency , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/physiology , Imaging, Three-Dimensional , Mathematical Concepts , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Models, Biological , Morphogenesis/genetics , Mutation , Phenotype , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphoproteins/physiology , Transforming Growth Factor beta2/deficiency , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/physiology
17.
J Pathol ; 248(4): 452-463, 2019 08.
Article in English | MEDLINE | ID: mdl-30916783

ABSTRACT

The establishment of the peristaltic machinery of the ureter is precisely controlled to cope with the onset of urine production in the fetal kidney. Retinoic acid (RA) has been identified as a signal that maintains the mesenchymal progenitors of the contractile smooth muscle cells (SMCs), while WNTs, SHH, and BMP4 induce their differentiation. How the activity of the underlying signalling pathways is controlled in time, space, and quantity to activate coordinately the SMC programme is poorly understood. Here, we provide evidence that the Zn-finger transcription factor GATA2 is involved in this crosstalk. In mice, Gata2 is expressed in the undifferentiated ureteric mesenchyme under control of RA signalling. Conditional deletion of Gata2 by a Tbx18cre driver results in hydroureter formation at birth, associated with a loss of differentiated SMCs. Analysis at earlier stages and in explant cultures revealed that SMC differentiation is not abrogated but delayed and that dilated ureters can partially regain peristaltic activity when relieved of urine pressure. Molecular analysis identified increased RA signalling as one factor contributing to the delay in SMC differentiation, possibly caused by reduced direct transcriptional activation of Cyp26a1, which encodes an RA-degrading enzyme. Our study identified GATA2 as a feedback inhibitor of RA signalling important for precise onset of ureteric SMC differentiation, and suggests that in a subset of cases of human congenital ureter dilatations, temporary relief of urine pressure may ameliorate the differentiation status of the SMC coat. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Cell Differentiation , GATA2 Transcription Factor/deficiency , Mesoderm/embryology , Myocytes, Smooth Muscle/physiology , Ureter/embryology , Ureteral Diseases/embryology , Animals , Biomarkers/metabolism , Female , GATA2 Transcription Factor/genetics , Male , Mesoderm/metabolism , Mice , Signal Transduction , Tretinoin/metabolism , Ureter/abnormalities , Ureter/metabolism , Ureteral Diseases/congenital , Ureteral Diseases/metabolism
18.
PLoS Genet ; 13(8): e1006951, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28797033

ABSTRACT

The differentiated cell types of the epithelial and mesenchymal tissue compartments of the mature ureter of the mouse arise in a precise temporal and spatial sequence from uncommitted precursor cells of the distal ureteric bud epithelium and its surrounding mesenchyme. Previous genetic efforts identified a member of the Hedgehog (HH) family of secreted proteins, Sonic hedgehog (SHH) as a crucial epithelial signal for growth and differentiation of the ureteric mesenchyme. Here, we used conditional loss- and gain-of-function experiments of the unique HH signal transducer Smoothened (SMO) to further characterize the cellular functions and unravel the effector genes of HH signaling in ureter development. We showed that HH signaling is not only required for proliferation and SMC differentiation of cells of the inner mesenchymal region but also for survival of cells of the outer mesenchymal region, and for epithelial proliferation and differentiation. We identified the Forkhead transcription factor gene Foxf1 as a target of HH signaling in the ureteric mesenchyme. Expression of a repressor version of FOXF1 in this tissue completely recapitulated the mesenchymal and epithelial proliferation and differentiation defects associated with loss of HH signaling while re-expression of a wildtype version of FOXF1 in the inner mesenchymal layer restored these cellular programs when HH signaling was inhibited. We further showed that expression of Bmp4 in the ureteric mesenchyme depends on HH signaling and Foxf1, and that exogenous BMP4 rescued cell proliferation and epithelial differentiation in ureters with abrogated HH signaling or FOXF1 function. We conclude that SHH uses a FOXF1-BMP4 module to coordinate the cellular programs for ureter elongation and differentiation, and suggest that deregulation of this signaling axis occurs in human congenital anomalies of the kidney and urinary tract (CAKUT).


Subject(s)
Bone Morphogenetic Protein 4/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Ureter/embryology , Animals , Bone Morphogenetic Protein 4/genetics , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Epithelium/embryology , Epithelium/metabolism , Female , Forkhead Transcription Factors/genetics , Hedgehog Proteins/genetics , Image Processing, Computer-Assisted , Male , Mesoderm/embryology , Mesoderm/metabolism , Mice , Microarray Analysis , Organogenesis/genetics , Reproducibility of Results , Signal Transduction , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Ureter/metabolism
19.
Proc Natl Acad Sci U S A ; 114(43): E9016-E9025, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073098

ABSTRACT

The elimination of unwanted cells by apoptosis is necessary for tissue morphogenesis. However, the cellular control of morphogenetic apoptosis is poorly understood, notably the modulation of cell sensitivity to apoptotic stimuli. Ureter maturation, the process by which the ureter is displaced to the bladder wall, represents an exquisite example of morphogenetic apoptosis, requiring the receptor protein tyrosine phosphatases (RPTPs): LAR and RPTPσ. Here we show that LAR-RPTPs act through cellular inhibitor of apoptosis protein 1 (cIAP1) to modulate caspase 3,7-mediated ureter maturation. Pharmacologic or genetic inactivation of cIAP1 reverts the apoptotic deficit of LAR-RPTP-deficient embryos. Moreover, Birc2 (cIAP1) inactivation generates excessive apoptosis leading to vesicoureteral reflux in newborns, which underscores the importance of apoptotic modulation during urinary tract morphogenesis. We finally demonstrate that LAR-RPTP deficiency increases cIAP1 stability during apoptotic cell death. Together these results identify a mode of cIAP1 regulation playing a critical role in the cellular response to apoptotic pathway activation in the embryo.


Subject(s)
Apoptosis/physiology , Gene Expression Regulation, Developmental/physiology , Inhibitor of Apoptosis Proteins/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Ureter/embryology , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cells, Cultured , Embryo, Mammalian , Fibroblasts/physiology , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, Knockout , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Signal Transduction , Ubiquitin-Protein Ligases , Ureter/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL