Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Hum Mol Genet ; 32(6): 1032-1047, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36282544

ABSTRACT

Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a congenital condition characterized by aplasia or hypoplasia of the uterus and vagina in women with a 46,XX karyotype. This condition can occur as type I when isolated or as type II when associated with extragenital anomalies including kidney and skeletal abnormalities. The genetic basis of MRKH syndrome remains unexplained and several candidate genes have been proposed to play a role in its etiology, including HNF1B, LHX1 and WNT4. Here, we conducted a microarray analysis of 13 women affected by MRKH syndrome, resulting in the identification of chromosomal changes, including the deletion at 17q12, which contains both HNF1B and LHX1. We focused on HNF1B for further investigation due to its known association with, but unknown etiological role in, MRKH syndrome. We ablated Hnf1b specifically in the epithelium of the Müllerian ducts in mice and found that this caused hypoplastic development of the uterus, as well as kidney anomalies, closely mirroring the MRKH type II phenotype. Using single-cell RNA sequencing of uterine tissue in the Hnf1b-ablated embryos, we analyzed the molecules and pathways downstream of Hnf1b, revealing a dysregulation of processes associated with cell proliferation, migration and differentiation. Thus, we establish that loss of Hnf1b function leads to an MRKH phenotype and generate the first mouse model of MRKH syndrome type II. Our results support the investigation of HNF1B in clinical genetic settings of MRKH syndrome and shed new light on the molecular mechanisms underlying this poorly understood condition in women's reproductive health.


Subject(s)
46, XX Disorders of Sex Development , Mullerian Ducts , Animals , Female , Mice , 46, XX Disorders of Sex Development/genetics , Cell Differentiation , Genomics , Hepatocyte Nuclear Factor 1-beta/genetics , Humans
2.
J Pathol ; 254(1): 31-45, 2021 05.
Article in English | MEDLINE | ID: mdl-33527355

ABSTRACT

Maturity-onset diabetes of the young type 5 (MODY5) is due to heterozygous mutations or deletion of HNF1B. No mouse models are currently available to recapitulate the human MODY5 disease. Here, we investigate the pancreatic phenotype of a unique MODY5 mouse model generated by heterozygous insertion of a human HNF1B splicing mutation at the intron-2 splice donor site in the mouse genome. This Hnf1bsp2/+ model generated with targeted mutation of Hnf1b mimicking the c.544+1G>T (T) mutation identified in humans, results in alternative transcripts and a 38% decrease of native Hnf1b transcript levels. As a clinical feature of MODY5 patients, the hypomorphic mouse model Hnf1bsp2/+ displays glucose intolerance. Whereas Hnf1bsp2/+ isolated islets showed no altered insulin secretion, we found a 65% decrease in pancreatic insulin content associated with a 30% decrease in total large islet volume and a 20% decrease in total ß-cell volume. These defects were associated with a 30% decrease in expression of the pro-endocrine gene Neurog3 that we previously identified as a direct target of Hnf1b, showing a developmental etiology. As another clinical feature of MODY5 patients, the Hnf1bsp2/+ pancreases display exocrine dysfunction with hypoplasia. We observed chronic pancreatitis with loss of acinar cells, acinar-to-ductal metaplasia, and lipomatosis, with upregulation of signaling pathways and impaired acinar cell regeneration. This was associated with ductal cell deficiency characterized by shortened primary cilia. Importantly, the Hnf1bsp2/+ mouse model reproduces the pancreatic features of the human MODY5/HNF1B disease, providing a unique in vivo tool for molecular studies of the endocrine and exocrine defects and to advance basic and translational research. © 2021 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)
Central Nervous System Diseases/genetics , Central Nervous System Diseases/physiopathology , Dental Enamel/abnormalities , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/physiopathology , Pancreas/physiopathology , Animals , Central Nervous System Diseases/pathology , Dental Enamel/pathology , Dental Enamel/physiopathology , Diabetes Mellitus, Type 2/pathology , Humans , Kidney Diseases, Cystic/pathology , Mice , Mice, Transgenic , Mutation , Pancreas/pathology , Phenotype
3.
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
4.
Development ; 144(6): 1113-1117, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28219945

ABSTRACT

Tissue, organ and organoid cultures provide suitable models for developmental studies, but our understanding of how the organs are assembled at the single-cell level still remains unclear. We describe here a novel fixed z-direction (FiZD) culture setup that permits high-resolution confocal imaging of organoids and embryonic tissues. In a FiZD culture a permeable membrane compresses the tissues onto a glass coverslip and the spacers adjust the thickness, enabling the tissue to grow for up to 12 days. Thus, the kidney rudiment and the organoids can adjust to the limited z-directional space and yet advance the process of kidney morphogenesis, enabling long-term time-lapse and high-resolution confocal imaging. As the data quality achieved was sufficient for computer-assisted cell segmentation and analysis, the method can be used for studying morphogenesis ex vivo at the level of the single constituent cells of a complex mammalian organogenesis model system.


Subject(s)
Kidney/embryology , Microscopy, Confocal/methods , Organoids/embryology , Time-Lapse Imaging/methods , Tissue Culture Techniques/methods , Animals , Imaging, Three-Dimensional , Mice , Morphogenesis
5.
Nature ; 494(7435): 111-5, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23389544

ABSTRACT

Insulin resistance represents a hallmark during the development of type 2 diabetes mellitus and in the pathogenesis of obesity-associated disturbances of glucose and lipid metabolism. MicroRNA (miRNA)-dependent post-transcriptional gene silencing has been recognized recently to control gene expression in disease development and progression, including that of insulin-resistant type 2 diabetes. The deregulation of miRNAs miR-143 (ref. 4), miR-181 (ref. 5), and miR-103 and miR-107 (ref. 6) alters hepatic insulin sensitivity. Here we report that the expression of miR-802 is increased in the liver of two obese mouse models and obese human subjects. Inducible transgenic overexpression of miR-802 in mice causes impaired glucose tolerance and attenuates insulin sensitivity, whereas reduction of miR-802 expression improves glucose tolerance and insulin action. We identify Hnf1b (also known as Tcf2) as a target of miR-802-dependent silencing, and show that short hairpin RNA (shRNA)-mediated reduction of Hnf1b in liver causes glucose intolerance, impairs insulin signalling and promotes hepatic gluconeogenesis. In turn, hepatic overexpression of Hnf1b improves insulin sensitivity in Lepr(db/db) mice. Thus, this study defines a critical role for deregulated expression of miR-802 in the development of obesity-associated impairment of glucose metabolism through targeting of Hnf1b, and assigns Hnf1b an unexpected role in the control of hepatic insulin sensitivity.


Subject(s)
Gene Silencing , Glucose/metabolism , Hepatocyte Nuclear Factor 1-beta/deficiency , MicroRNAs/genetics , Obesity/genetics , Animals , Gene Expression Regulation , Gluconeogenesis , Glucose/biosynthesis , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Humans , Insulin/metabolism , Insulin Resistance/genetics , Liver/metabolism , Mice , MicroRNAs/biosynthesis , Signal Transduction
6.
J Proteome Res ; 17(7): 2293-2306, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29873499

ABSTRACT

In the present study, we performed a metabolomics analysis to evaluate a MODY5/RCAD mouse mutant line as a potential model for HNF1B-associated diseases. Gas chromatography time-of-flight mass spectrometry (GC-TOF-MS) of gut, kidney, liver, muscle, pancreas, and plasma samples uncovered the tissue specific metabolite distribution. Orthogonal projections to latent structures discriminant analysis (OPLS-DA) was used to identify the differences between MODY5/RCAD and wild-type mice in each of the tissues. The differences included, for example, increased levels of amino acids in the kidneys and reduced levels of fatty acids in the muscles of the MODY5/RCAD mice. Interestingly, campesterol was found in higher concentrations in the MODY5/RCAD mice, with a four-fold and three-fold increase in kidneys and pancreas, respectively. As expected, the MODY5/RCAD mice displayed signs of impaired renal function in addition to disturbed liver lipid metabolism, with increased lipid and fatty acid accumulation in the liver. From a metabolomics perspective, the MODY5/RCAD model was proven to display a metabolic pattern similar to what would be suspected in HNF1B-associated diseases. These findings were in line with the presumed outcome of the mutation based on the different anatomy and function of the tissues as well as the effect of the mutation on development.


Subject(s)
Disease Models, Animal , Metabolomics/methods , Mice, Mutant Strains/metabolism , Animals , Cadherins/genetics , Gas Chromatography-Mass Spectrometry , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney/metabolism , Liver/metabolism , Mice , Pancreas/metabolism
7.
Development ; 142(5): 871-82, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25715395

ABSTRACT

Heterozygous mutations in the human HNF1B gene are associated with maturity-onset diabetes of the young type 5 (MODY5) and pancreas hypoplasia. In mouse, Hnf1b heterozygous mutants do not exhibit any phenotype, whereas the homozygous deletion in the entire epiblast leads to pancreas agenesis associated with abnormal gut regionalization. Here, we examine the specific role of Hnf1b during pancreas development, using constitutive and inducible conditional inactivation approaches at key developmental stages. Hnf1b early deletion leads to a reduced pool of pancreatic multipotent progenitor cells (MPCs) due to decreased proliferation and increased apoptosis. Lack of Hnf1b either during the first or the secondary transitions is associated with cystic ducts. Ductal cells exhibit aberrant polarity and decreased expression of several cystic disease genes, some of which we identified as novel Hnf1b targets. Notably, we show that Glis3, a transcription factor involved in duct morphogenesis and endocrine cell development, is downstream Hnf1b. In addition, a loss and abnormal differentiation of acinar cells are observed. Strikingly, inactivation of Hnf1b at different time points results in the absence of Ngn3(+) endocrine precursors throughout embryogenesis. We further show that Hnf1b occupies novel Ngn3 putative regulatory sequences in vivo. Thus, Hnf1b plays a crucial role in the regulatory networks that control pancreatic MPC expansion, acinar cell identity, duct morphogenesis and generation of endocrine precursors. Our results uncover an unappreciated requirement of Hnf1b in endocrine cell specification and suggest a mechanistic explanation of diabetes onset in individuals with MODY5.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hepatocyte Nuclear Factor 1-beta/metabolism , Nerve Tissue Proteins/metabolism , Pancreas/cytology , Pancreas/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/physiology , Chromatin Immunoprecipitation , Cystic Duct/cytology , Cystic Duct/metabolism , DNA-Binding Proteins , Female , Hepatocyte Nuclear Factor 1-beta/genetics , Immunohistochemistry , In Situ Nick-End Labeling , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Pregnancy , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
8.
Development ; 140(4): 873-85, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23362348

ABSTRACT

The nephron is a highly specialised segmented structure that provides essential filtration and resorption renal functions. It arises by formation of a polarised renal vesicle that differentiates into a comma-shaped body and then a regionalised S-shaped body (SSB), with the main prospective segments mapped to discrete domains. The regulatory circuits involved in initial nephron patterning are poorly understood. We report here that HNF1B, a transcription factor known to be involved in ureteric bud branching and initiation of nephrogenesis, has an additional role in segment fate acquisition. Hnf1b conditional inactivation in murine nephron progenitors results in rudimentary nephrons comprising a glomerulus connected to the collecting system by a short tubule displaying distal fates. Renal vesicles develop and polarise normally but fail to progress to correctly patterned SSBs. Major defects are evident at late SSBs, with altered morphology, reduction of a proximo-medial subdomain and increased apoptosis. This is preceded by strong downregulation of the Notch pathway components Lfng, Dll1 and Jag1 and the Irx1/2 factors, which are potential regulators of proximal and Henle's loop segment fates. Moreover, HNF1B is recruited to the regulatory sequences of most of these genes. Overexpression of a HNF1B dominant-negative construct in Xenopus embryos causes downregulation specifically of proximal and intermediate pronephric segment markers. These results show that HNF1B is required for the acquisition of a proximo-intermediate segment fate in vertebrates, thus uncovering a previously unappreciated function of a novel SSB subcompartment in global nephron segmentation and further differentiation.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Hepatocyte Nuclear Factor 1-beta/metabolism , Nephrons/embryology , Organogenesis/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Chromatin Immunoprecipitation , Gene Expression Regulation, Developmental/genetics , Histological Techniques , Homeodomain Proteins/metabolism , Imaging, Three-Dimensional , Immunohistochemistry , In Situ Hybridization , In Situ Nick-End Labeling , Mice , Nephrons/metabolism , Organogenesis/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tomography, Optical , Transcription Factors/metabolism
9.
J Biol Chem ; 288(23): 16655-16670, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23592779

ABSTRACT

Cubilin (Cubn) is a multiligand endocytic receptor critical for the intestinal absorption of vitamin B12 and renal protein reabsorption. During mouse development, Cubn is expressed in both embryonic and extra-embryonic tissues, and Cubn gene inactivation results in early embryo lethality most likely due to the impairment of the function of extra-embryonic Cubn. Here, we focus on the developmental role of Cubn expressed in the embryonic head. We report that Cubn is a novel, interspecies-conserved Fgf receptor. Epiblast-specific inactivation of Cubn in the mouse embryo as well as Cubn silencing in the anterior head of frog or the cephalic neural crest of chick embryos show that Cubn is required during early somite stages to convey survival signals in the developing vertebrate head. Surface plasmon resonance analysis reveals that fibroblast growth factor 8 (Fgf8), a key mediator of cell survival, migration, proliferation, and patterning in the developing head, is a high affinity ligand for Cubn. Cell uptake studies show that binding to Cubn is necessary for the phosphorylation of the Fgf signaling mediators MAPK and Smad1. Although Cubn may not form stable ternary complexes with Fgf receptors (FgfRs), it acts together with and/or is necessary for optimal FgfR activity. We propose that plasma membrane binding of Fgf8, and most likely of the Fgf8 family members Fgf17 and Fgf18, to Cubn improves Fgf ligand endocytosis and availability to FgfRs, thus modulating Fgf signaling activity.


Subject(s)
Fibroblast Growth Factor 8/metabolism , Head/embryology , MAP Kinase Signaling System/physiology , Neural Crest/embryology , Receptors, Cell Surface/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Animals , Cell Survival/physiology , Endocytosis/physiology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Silencing , Ligands , Mice , Mice, Transgenic , Neural Crest/cytology , Protein Binding , Receptors, Cell Surface/genetics , Receptors, Fibroblast Growth Factor/genetics
10.
Hum Mol Genet ; 21(14): 3143-55, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22511595

ABSTRACT

The transcription factors HNF1B and Pax2, co-expressed in the Wolffian duct and ureteric bud epithelia, play essential roles during the early steps of mouse kidney development. In humans, heterozygous mutations in these genes display a number of common kidney phenotypes, including hypoplasia and multicystic hypoplastic kidneys. Moreover, a high prevalence of mutations either in HNF1B or PAX2 has been observed in children with renal hypodysplasia. To gain a better understanding of Hnf1b and Pax2 interactions in vivo, we generated compound heterozygous mice for Hnf1b and Pax2 null alleles. We show here that compound heterozygous mutants display phenotypes similar to severe congenital anomalies of the kidney and the urinary tract (CAKUT), including strong hypoplasia of the kidneys, caudal ectopic aborted ureter buds, duplex kidneys, megaureters and hydronephrosis. At a molecular level, compound mutants show a delay in nephron segment and medullar interstitial differentiation, increased apoptosis and a transient decrease in Lim1 and Wnt4 expression. We also observe a perturbation of smooth muscle differentiation around the ureter associated with a local down-regulation in transcript levels of Bmp4 and Tbx18, two key regulators involved in ureter smooth muscle formation, thus explaining, at least in part, megaureters. These results together uncover a novel role of Hnf1b as a modifier of the Pax2 haplo-insufficient phenotype and show that these two transcription factors operate in common pathways governing both kidney morphogenesis and ureter differentiation. This mouse model should provide new insights into the pathogenic mechanisms of human CAKUT, the most frequent developmental defect identified in newborns.


Subject(s)
Hepatocyte Nuclear Factor 1-beta/metabolism , Kidney Diseases/metabolism , Kidney/metabolism , Morphogenesis , PAX2 Transcription Factor/metabolism , Ureter/metabolism , Ureteral Diseases/metabolism , Animals , Disease Models, Animal , Female , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Kidney/embryology , Kidney Diseases/embryology , Kidney Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PAX2 Transcription Factor/genetics , Ureter/embryology , Ureteral Diseases/embryology , Ureteral Diseases/genetics
11.
Stem Cell Reports ; 19(6): 859-876, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38788724

ABSTRACT

Hepatocyte nuclear factor 1B (HNF1B) encodes a transcription factor expressed in developing human kidney epithelia. Heterozygous HNF1B mutations are the commonest monogenic cause of dysplastic kidney malformations (DKMs). To understand their pathobiology, we generated heterozygous HNF1B mutant kidney organoids from CRISPR-Cas9 gene-edited human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) reprogrammed from a family with HNF1B-associated DKMs. Mutant organoids contained enlarged malformed tubules displaying deregulated cell turnover. Numerous genes implicated in Mendelian kidney tubulopathies were downregulated, and mutant tubules resisted the cyclic AMP (cAMP)-mediated dilatation seen in controls. Bulk and single-cell RNA sequencing (scRNA-seq) analyses indicated abnormal Wingless/Integrated (WNT), calcium, and glutamatergic pathways, the latter hitherto unstudied in developing kidneys. Glutamate ionotropic receptor kainate type subunit 3 (GRIK3) was upregulated in malformed mutant nephron tubules and prominent in HNF1B mutant fetal human dysplastic kidney epithelia. These results reveal morphological, molecular, and physiological roles for HNF1B in human kidney tubule differentiation and morphogenesis illuminating the developmental origin of mutant-HNF1B-causing kidney disease.


Subject(s)
Hepatocyte Nuclear Factor 1-beta , Induced Pluripotent Stem Cells , Organoids , Humans , Hepatocyte Nuclear Factor 1-beta/genetics , Hepatocyte Nuclear Factor 1-beta/metabolism , Organoids/metabolism , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , Heterozygote , Kidney Tubules/pathology , Kidney Tubules/metabolism , Mutation , Kidney/pathology , Kidney/metabolism , Kidney/abnormalities , CRISPR-Cas Systems , Pluripotent Stem Cells/metabolism , Gene Editing
12.
Development ; 137(2): 347-57, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20040500

ABSTRACT

Mouse metanephric kidney development begins with the induction of the ureteric bud (UB) from the caudal portion of the Wolffian duct by metanephric mesenchymal signals. While the UB undergoes branching morphogenesis to generate the entire urinary collecting system and the ureter, factors secreted by the UB tips induce surrounding mesenchymal cells to convert into epithelia and form the nephrons, the functional units of the kidney. Epithelial branching morphogenesis and nephrogenesis are therefore tightly orchestrated; defects in either of these processes lead to severe kidney phenotypes ranging from hypoplasia to complete aplasia. However, the underlying regulatory networks have been only partially elucidated. Here, we identify the transcription factor vHNF1 (HNF1beta) as a crucial regulator of these early developmental events. Initially involved in timing outgrowth of the UB and subsequent branching, vHNF1 is also required for nephric duct epithelial maintenance, Müllerian duct formation and early nephrogenesis. Mosaic analyses further suggest a cell-autonomous requirement for vHNF1 in the acquisition of a specialized tip domain and branching morphogenesis. vHNF1 exerts these intricate functions at least in part through the direct control of key regulatory molecules involved in different aspects of early kidney development. Notably, vHNF1 acting directly upstream of Wnt9b appears to orchestrate Wnt signaling action in the mesenchymal-epithelial transitions underlying the initiation of nephrogenesis. These results demonstrate that vHNF1 is an essential transcriptional regulator that, in addition to the known later functions in normal duct morphogenesis, plays a crucial role during the earliest stages of urogenital development and provide novel insights into the regulatory circuits controlling events.


Subject(s)
Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-beta/metabolism , Kidney/embryology , Organogenesis/physiology , Ureter/embryology , Animals , Cell Line , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Hepatocyte Nuclear Factor 1-beta/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Kidney/metabolism , Mice , Organogenesis/genetics , Reverse Transcriptase Polymerase Chain Reaction , Ureter/metabolism
13.
Dis Model Mech ; 14(5)2021 05 01.
Article in English | MEDLINE | ID: mdl-33737325

ABSTRACT

Heterozygous mutations in HNF1B cause the complex syndrome renal cysts and diabetes (RCAD), characterized by developmental abnormalities of the kidneys, genital tracts and pancreas, and a variety of renal, pancreas and liver dysfunctions. The pathogenesis underlying this syndrome remains unclear as mice with heterozygous null mutations have no phenotype, while constitutive/conditional Hnf1b ablation leads to more severe phenotypes. We generated a novel mouse model carrying an identified human mutation at the intron-2 splice donor site. Unlike heterozygous mice previously characterized, mice heterozygous for the splicing mutation exhibited decreased HNF1B protein levels and bilateral renal cysts from embryonic day 15, originated from glomeruli, early proximal tubules (PTs) and intermediate nephron segments, concurrently with delayed PT differentiation, hydronephrosis and rare genital tract anomalies. Consistently, mRNA sequencing showed that most downregulated genes in embryonic kidneys were primarily expressed in early PTs and the loop of Henle and involved in ion/drug transport, organic acid and lipid metabolic processes, while the expression of previously identified targets upon Hnf1b ablation, including cystic disease genes, was weakly or not affected. Postnatal analyses revealed renal abnormalities, ranging from glomerular cysts to hydronephrosis and, rarely, multicystic dysplasia. Urinary proteomics uncovered a particular profile predictive of progressive decline in kidney function and fibrosis, and displayed common features with a recently reported urine proteome in an RCAD pediatric cohort. Altogether, our results show that reduced HNF1B levels lead to developmental disease phenotypes associated with the deregulation of a subset of HNF1B targets. They further suggest that this model represents a unique clinical/pathological viable model of the RCAD disease.


Subject(s)
Central Nervous System Diseases/genetics , Dental Enamel/abnormalities , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Genes, Developmental , Haploinsufficiency/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Kidney Diseases, Cystic/genetics , Animals , Animals, Newborn , Cell Polarity , Central Nervous System Diseases/pathology , Cilia/pathology , Dental Enamel/pathology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Embryo, Mammalian/pathology , Gene Dosage , Gene Expression Profiling , Heterozygote , Humans , Hydronephrosis/complications , Kidney Diseases, Cystic/pathology , Kidney Glomerulus/pathology , Kidney Tubules/pathology , Mice, Inbred C57BL , Mutation/genetics , Nephrons/pathology , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Severity of Illness Index
14.
Cell Mol Gastroenterol Hepatol ; 8(3): 487-511, 2019.
Article in English | MEDLINE | ID: mdl-31229598

ABSTRACT

BACKGROUND & AIMS: The exocrine pancreas consists of acinar cells that produce digestive enzymes transported to the intestine through a branched ductal epithelium. Chronic pancreatitis is characterized by progressive inflammation, fibrosis, and loss of acinar tissue. These changes of the exocrine tissue are risk factors for pancreatic cancer. The cause of chronic pancreatitis cannot be identified in one quarter of patients. Here, we investigated how duct dysfunction could contribute to pancreatitis development. METHODS: The transcription factor Hnf1b, first expressed in pancreatic progenitors, is strictly restricted to ductal cells from late embryogenesis. We previously showed that Hnf1b is crucial for pancreas morphogenesis but its postnatal role still remains unelucidated. To investigate the role of pancreatic ducts in exocrine homeostasis, we inactivated the Hnf1b gene in vivo in mouse ductal cells. RESULTS: We uncovered that postnatal Hnf1b inactivation in pancreatic ducts leads to chronic pancreatitis in adults. Hnf1bΔduct mutants show dilatation of ducts, loss of acinar cells, acinar-to-ductal metaplasia, and lipomatosis. We deciphered the early events involved, with down-regulation of cystic disease-associated genes, loss of primary cilia, up-regulation of signaling pathways, especially the Yap pathway, which is involved in acinar-to-ductal metaplasia. Remarkably, Hnf1bΔduct mutants developed pancreatic intraepithelial neoplasia and promote pancreatic intraepithelial neoplasia progression in concert with KRAS. We further showed that adult Hnf1b inactivation in pancreatic ducts is associated with impaired regeneration after injury, with persistent metaplasia and initiation of neoplasia. CONCLUSIONS: Loss of Hnf1b in ductal cells leads to chronic pancreatitis and neoplasia. This study shows that Hnf1b deficiency may contribute to diseases of the exocrine pancreas and gains further insight into the etiology of pancreatitis and tumorigenesis.


Subject(s)
Carcinoma in Situ/genetics , Gene Deletion , Hepatocyte Nuclear Factor 1-beta/genetics , Pancreatic Ducts/growth & development , Pancreatic Neoplasms/genetics , Pancreatitis/genetics , Animals , Animals, Newborn , Carcinoma in Situ/metabolism , Female , Genetic Predisposition to Disease , Hepatocyte Nuclear Factor 1-beta/metabolism , Homeostasis , Humans , Mice , Pancreas, Exocrine/metabolism , Pancreatic Ducts/metabolism , Pancreatic Neoplasms/metabolism , Pancreatitis/complications , Pancreatitis/metabolism , Signal Transduction
15.
Sci Rep ; 9(1): 2225, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30778115

ABSTRACT

Renal Cysts and Diabetes Syndrome (RCAD) is an autosomal dominant disorder caused by mutations in the HNF1B gene encoding for the transcriptional factor hepatocyte nuclear factor-1B. RCAD is characterized as a multi-organ disease, with a broad spectrum of symptoms including kidney abnormalities (renal cysts, renal hypodysplasia, single kidney, horseshoe kidneys, hydronephrosis), early-onset diabetes mellitus, abnormal liver function, pancreatic hypoplasia and genital tract malformations. In the present study, using capillary electrophoresis coupled to mass spectrometry (CE-MS), we investigated the urinary proteome of a pediatric cohort of RCAD patients and different controls to identify peptide biomarkers and obtain further insights into the pathophysiology of this disorder. As a result, 146 peptides were found to be associated with RCAD in 22 pediatric patients when compared to 22 healthy age-matched controls. A classifier based on these peptides was generated and further tested on an independent cohort, clearly discriminating RCAD patients from different groups of controls. This study demonstrates that the urinary proteome of pediatric RCAD patients differs from autosomal dominant polycystic kidney disease (PKD1, PKD2), congenital nephrotic syndrome (NPHS1, NPHS2, NPHS4, NPHS9) as well as from chronic kidney disease conditions, suggesting differences between the pathophysiology behind these disorders.


Subject(s)
Biomarkers , Central Nervous System Diseases/metabolism , Dental Enamel/abnormalities , Diabetes Mellitus, Type 2/metabolism , Kidney Diseases, Cystic/metabolism , Proteome , Proteomics , Adolescent , Biomarkers/urine , Central Nervous System Diseases/diagnosis , Central Nervous System Diseases/urine , Child , Child, Preschool , Dental Enamel/metabolism , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/urine , Diagnosis, Differential , Female , Humans , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/urine , Male , Mass Spectrometry , Peptides/urine , Phenotype , Proteomics/methods , Reproducibility of Results
16.
Diabetes ; 55(1): 61-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16380477

ABSTRACT

Generation of pancreatic precursor cells in the endoderm is controlled by a network of transcription factors. Hepatocyte nuclear factor-6 (HNF6) is a key player in this network, because it controls the initiation of the expression of pancreatic and duodenal homeobox 1 (Pdx1), the earliest marker of pancreatic precursor cells. To further characterize this network, we have investigated how the expression of HNF6 is controlled in mouse endoderm, by using in vitro and in vivo protein-DNA interaction techniques combined with endoderm electroporation, transgenesis, and gene inactivation in embryos. We delineated Hnf6 regulatory regions that confer expression of a reporter gene in the embryonic endoderm but not in extraembryonic visceral endoderm. HNF6 expression in the embryonic endoderm was found to depend on an intronic enhancer. This enhancer contains functional binding sites for the tissue-specific factors of the forkhead box A and HNF1 families. Among the latter, variant HNF1 (vHNF1)/TCF2, which is expressed before HNF6 in the endoderm, was found to be critical for HNF6 expression. Therefore, the sequential activation of vHNF1, HNF6, and Pdx1 in the endoderm appears to control the generation of pancreatic precursors. This cascade may be used to benchmark in vitro differentiation of pancreatic precursor cells from embryonic stem cells, for cell therapy of diabetes.


Subject(s)
Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocyte Nuclear Factor 1-beta/metabolism , Hepatocyte Nuclear Factor 6/genetics , Hepatocyte Nuclear Factor 6/metabolism , Pancreas/cytology , Pancreas/embryology , Animals , Chimera , Endoderm/metabolism , Enhancer Elements, Genetic/genetics , Gene Deletion , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Introns/genetics , Mice , Promoter Regions, Genetic/genetics , Trans-Activators/genetics , Trans-Activators/metabolism
17.
Mol Cell Biol ; 24(21): 9508-16, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15485918

ABSTRACT

The mammalian target of rapamycin (mTOR) is a key component of a signaling pathway which integrates inputs from nutrients and growth factors to regulate cell growth. Recent studies demonstrated that mice harboring an ethylnitrosourea-induced mutation in the gene encoding mTOR die at embryonic day 12.5 (E12.5). However, others have shown that the treatment of E4.5 blastocysts with rapamycin blocks trophoblast outgrowth, suggesting that the absence of mTOR should lead to embryonic lethality at an earlier stage. To resolve this discrepancy, we set out to disrupt the mTOR gene and analyze the outcome in both heterozygous and homozygous settings. Heterozygous mTOR (mTOR(+/-)) mice do not display any overt phenotype, although mouse embryonic fibroblasts derived from these mice show a 50% reduction in mTOR protein levels and phosphorylation of S6 kinase 1 T389, a site whose phosphorylation is directly mediated by mTOR. However, S6 phosphorylation, raptor levels, cell size, and cell cycle transit times are not diminished in these cells. In contrast to the situation in mTOR(+/-) mice, embryonic development of homozygous mTOR(-/-) mice appears to be arrested at E5.5; such embryos are severely runted and display an aberrant developmental phenotype. The ability of these embryos to implant corresponds to a limited level of trophoblast outgrowth in vitro, reflecting a maternal mRNA contribution, which has been shown to persist during preimplantation development. Moreover, mTOR(-/-) embryos display a lesion in inner cell mass proliferation, consistent with the inability to establish embryonic stem cells from mTOR(-/-) embryos.


Subject(s)
Embryo Loss/genetics , Embryo Loss/pathology , Embryonic Development/physiology , Protein Kinases/deficiency , Protein Kinases/metabolism , Stem Cells/metabolism , Stem Cells/pathology , Alleles , Animals , Cell Differentiation , Cell Division , Cell Size , Cells, Cultured , Female , Fibroblasts , Gene Deletion , Heterozygote , Mice , Mice, Knockout , Pregnancy , Protein Kinases/genetics , TOR Serine-Threonine Kinases
18.
Cells ; 4(3): 483-99, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26378582

ABSTRACT

The nephron is the basic structural and functional unit of the vertebrate kidney. To ensure kidney functions, the nephrons possess a highly segmental organization where each segment is specialized for the secretion and reabsorption of particular solutes. During embryogenesis, nephron progenitors undergo a mesenchymal-to-epithelial transition (MET) and acquire different segment-specific cell fates along the proximo-distal axis of the nephron. Even if the morphological changes occurring during nephrogenesis are characterized, the regulatory networks driving nephron segmentation are still poorly understood. Interestingly, several studies have shown that the pronephric nephrons in Xenopus and zebrafish are segmented in a similar fashion as the mouse metanephric nephrons. Here we review functional and molecular aspects of nephron segmentation with a particular interest on the signaling molecules and transcription factors recently implicated in kidney development in these three different vertebrate model organisms. A complete understanding of the mechanisms underlying nephrogenesis in different model organisms will provide novel insights on the etiology of several human renal diseases.

19.
PLoS One ; 10(6): e0129260, 2015.
Article in English | MEDLINE | ID: mdl-26086868

ABSTRACT

Hierarchical modelling was applied in order to identify the organs that contribute to the levels of metabolites in plasma. Plasma and organ samples from gut, kidney, liver, muscle and pancreas were obtained from mice. The samples were analysed using gas chromatography time-of-flight mass spectrometry (GC TOF-MS) at the Swedish Metabolomics centre, Umeå University, Sweden. The multivariate analysis was performed by means of principal component analysis (PCA) and orthogonal projections to latent structures (OPLS). The main goal of this study was to investigate how each organ contributes to the metabolic plasma profile. This was performed using hierarchical modelling. Each organ was found to have a unique metabolic profile. The hierarchical modelling showed that the gut, kidney and liver demonstrated the greatest contribution to the metabolic pattern of plasma. For example, we found that metabolites were absorbed in the gut and transported to the plasma. The kidneys excrete branched chain amino acids (BCAAs) and fatty acids are transported in the plasma to the muscles and liver. Lactic acid was also found to be transported from the pancreas to plasma. The results indicated that hierarchical modelling can be utilized to identify the organ contribution of unknown metabolites to the metabolic profile of plasma.


Subject(s)
Gastrointestinal Tract/metabolism , Kidney/metabolism , Liver/metabolism , Metabolome , Metabolomics/methods , Models, Theoretical , Muscle, Skeletal/metabolism , Pancreas/metabolism , Animals , Gas Chromatography-Mass Spectrometry/methods , Mice
20.
Methods Mol Biol ; 886: 275-91, 2012.
Article in English | MEDLINE | ID: mdl-22639270

ABSTRACT

Chromatin immunoprecipitation (ChIP) is a powerful technique for examining transcription factor recruitment to chromatin, or histone modifications, at the level of specific genomic sequences. As such, it provides an invaluable tool for elucidating gene regulation at the molecular level. Combined with high-throughput methods such as second generation sequencing (ChIP-Seq), this technique is now commonly used for studying DNA-protein interactions at a genome-wide scale. The ChIP technique is based on covalent cross-linking of DNA and proteins with formaldehyde, followed by chromatin fragmentation, either enzymatic or by sonication, and immunoprecipitation of protein-DNA complexes using antibodies specific for the protein of interest. The immunoprecipitated DNA is then purified and the DNA sequences associated with the immunoprecipitated protein are identified by PCR (ChIP-PCR) or, alternatively, by direct sequencing (ChIP-Seq). Initially, the vast majority of ChIP experiments were performed on cultured cell lines. More recently, this technique has been adapted to a variety of tissues in different model organisms. We describe here a ChIP protocol on freshly isolated mouse embryonic kidneys for in vivo analysis of transcription factor recruitment on chromatin. This protocol has been easily adapted to other mouse embryonic tissues and has also been successfully scaled up to perform ChIP-Seq.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/chemistry , Kidney/embryology , Transcription Factors/analysis , Animals , DNA/genetics , DNA/isolation & purification , Dissection/methods , Kidney/surgery , Mice , Polymerase Chain Reaction/methods , Sequence Analysis, DNA/methods , Sonication/methods
SELECTION OF CITATIONS
SEARCH DETAIL