Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
JCI Insight ; 8(3)2023 02 08.
Article in English | MEDLINE | ID: mdl-36626229

ABSTRACT

Preterm birth results in low nephron endowment and increased risk of acute kidney injury (AKI) and chronic kidney disease (CKD). To understand the pathogenesis of AKI and CKD in preterm humans, we generated potentially novel mouse models with a 30%-70% reduction in nephron number by inhibiting or deleting Ret tyrosine kinase in the developing ureteric bud. These mice developed glomerular and tubular hypertrophy, followed by the transition to CKD, recapitulating the renal pathological changes seen in humans born preterm. We injected neonatal mice with gentamicin, a ubiquitous nephrotoxic exposure in preterm infants, and detected more severe proximal tubular injury in mice with low nephron number compared with controls with normal nephron number. Mice with low nephron number had reduced proliferative repair with more rapid development of CKD. Furthermore, mice had more profound inflammation with highly elevated levels of MCP-1 and CXCL10, produced in part by damaged proximal tubules. Our study directly links low nephron endowment with postnatal renal hypertrophy, which in this model is maladaptive and results in CKD. Underdeveloped kidneys are more susceptible to gentamicin-induced AKI, suggesting that AKI in the setting of low nephron number is more severe and further increases the risk of CKD in this vulnerable population.


Subject(s)
Acute Kidney Injury , Premature Birth , Renal Insufficiency, Chronic , Animals , Female , Humans , Mice , Acute Kidney Injury/pathology , Gentamicins , Hypertrophy/pathology , Infant, Premature , Kidney/pathology , Nephrons/pathology , Premature Birth/pathology , Renal Insufficiency, Chronic/pathology
2.
Development ; 148(9)2021 05 01.
Article in English | MEDLINE | ID: mdl-33914865

ABSTRACT

Ret signaling promotes branching morphogenesis during kidney development, but the underlying cellular mechanisms remain unclear. While Ret-expressing progenitor cells proliferate at the ureteric bud tips, some of these cells exit the tips to generate the elongating collecting ducts, and in the process turn off Ret. Genetic ablation of Ret in tip cells promotes their exit, suggesting that Ret is required for cell rearrangements that maintain the tip compartments. Here, we examine the behaviors of ureteric bud cells that are genetically forced to maintain Ret expression. These cells move to the nascent tips, and remain there during many cycles of branching; this tip-seeking behavior may require positional signals from the mesenchyme, as it occurs in whole kidneys but not in epithelial ureteric bud organoids. In organoids, cells forced to express Ret display a striking self-organizing behavior, attracting each other to form dense clusters within the epithelium, which then evaginate to form new buds. The ability of forced Ret expression to promote these events suggests that similar Ret-dependent cell behaviors play an important role in normal branching morphogenesis.


Subject(s)
Cell Movement , Epithelial Cells/metabolism , Signal Transduction , Ureter/metabolism , Animals , Cluster Analysis , Epithelium/metabolism , Female , Kidney/growth & development , Kidney/metabolism , Male , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Morphogenesis , Organoids , Protein-Tyrosine Kinases/metabolism , Stem Cells/metabolism
3.
Methods Mol Biol ; 1926: 3-21, 2019.
Article in English | MEDLINE | ID: mdl-30742258

ABSTRACT

For studies of gene function during development, it can be very useful to generate mosaic embryos in which a small subset of cells in a given cell lineage lacks a gene of interest and carries a marker that allows the mutant cells to be specifically visualized and compared to wild-type cells. Several methods have been used to generate genetically mosaic mouse kidneys for such studies. These include (1) chimeric embryos generated using embryonic stem cells, (2) chimeric renal organoids generated by dissociation and reaggregation of the fetal kidneys, (3) generation of a knockout allele with a built-in reporter gene, (4) mosaic analysis with double markers (MADM), and (5) mosaic mutant analysis with spatial and temporal control of recombination (MASTR). In this chapter, these five methods are described, and their advantages and disadvantages are discussed.


Subject(s)
Kidney/cytology , Kidney/embryology , Organoids/cytology , Animals , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Integrases/genetics , Integrases/metabolism , Kidney/metabolism , Mice , Mice, Knockout , Organoids/metabolism
4.
PLoS Biol ; 14(2): e1002382, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26894589

ABSTRACT

Branching morphogenesis of the epithelial ureteric bud forms the renal collecting duct system and is critical for normal nephron number, while low nephron number is implicated in hypertension and renal disease. Ureteric bud growth and branching requires GDNF signaling from the surrounding mesenchyme to cells at the ureteric bud tips, via the Ret receptor tyrosine kinase and coreceptor Gfrα1; Ret signaling up-regulates transcription factors Etv4 and Etv5, which are also critical for branching. Despite extensive knowledge of the genetic control of these events, it is not understood, at the cellular level, how renal branching morphogenesis is achieved or how Ret signaling influences epithelial cell behaviors to promote this process. Analysis of chimeric embryos previously suggested a role for Ret signaling in promoting cell rearrangements in the nephric duct, but this method was unsuited to study individual cell behaviors during ureteric bud branching. Here, we use Mosaic Analysis with Double Markers (MADM), combined with organ culture and time-lapse imaging, to trace the movements and divisions of individual ureteric bud tip cells. We first examine wild-type clones and then Ret or Etv4 mutant/wild-type clones in which the mutant and wild-type sister cells are differentially and heritably marked by green and red fluorescent proteins. We find that, in normal kidneys, most individual tip cells behave as self-renewing progenitors, some of whose progeny remain at the tips while others populate the growing UB trunks. In Ret or Etv4 MADM clones, the wild-type cells generated at a UB tip are much more likely to remain at, or move to, the new tips during branching and elongation, while their Ret-/- or Etv4-/- sister cells tend to lag behind and contribute only to the trunks. By tracking successive mitoses in a cell lineage, we find that Ret signaling has little effect on proliferation, in contrast to its effects on cell movement. Our results show that Ret/Etv4 signaling promotes directed cell movements in the ureteric bud tips, and suggest a model in which these cell movements mediate branching morphogenesis.


Subject(s)
Kidney/embryology , Morphogenesis , Proto-Oncogene Proteins c-ets/physiology , Proto-Oncogene Proteins c-ret/physiology , Stem Cells/physiology , Animals , Cell Movement , Female , Male , Mice , Organ Culture Techniques
5.
Dev Dyn ; 245(4): 483-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26813041

ABSTRACT

BACKGROUND: Cell rearrangements mediated by GDNF/Ret signaling underlie the formation of the ureteric bud (UB) tip domain during kidney development. Whether FGF signaling also influences these rearrangements is unknown. Chimeric embryos are a powerful tool for examining the genetic controls of cellular behaviors, but generating chimeras by traditional methods is expensive and laborious. Dissociated fetal kidney cells can reorganize to form complex structures including branching UB tubules, providing an easier method to generate renal chimeras. RESULTS: Cell behaviors in normal or chimeric kidney cultures were investigated using time-lapse imaging. In Spry1(-/-) ↔ wild-type chimeras, cells lacking Spry1 (a negative regulator of Ret and FGF receptor signaling) preferentially occupied the UB tips, as previously observed in traditional chimeras, thus validating this experimental system. In Fgfr2(UB-/-) ↔ wild-type chimeras, the wild-type cells preferentially occupied the tips. Independent evidence for a role of Fgfr2 in UB tip formation was obtained using Mosaic mutant Analysis with Spatial and Temporal control of Recombination (MASTR). CONCLUSIONS: Dissociation and reaggregation of fetal kidney cells of different genotypes, with suitable fluorescent markers, provides an efficient way to analyze cell behaviors in chimeric cultures. FGF/Fgfr2 signaling promotes UB cell rearrangements that form the tip domain, similarly to GDNF/Ret signaling.


Subject(s)
Kidney/cytology , Kidney/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Mice , Mice, Knockout
6.
J Biol Chem ; 290(37): 22460-73, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26224636

ABSTRACT

The pluripotency and self-renewal capacity of embryonic stem (ES) cells is regulated by several transcription factors. Here, we show that the ETS-related transcription factors Etv4 and Etv5 (Etv4/5) are specifically expressed in undifferentiated ES cells, and suppression of Oct3/4 results in down-regulation of Etv4/5. Simultaneous deletion of Etv4 and Etv5 (Etv4/5 double knock-out (dKO)) in ES cells resulted in a flat, epithelial cell-like appearance, whereas the morphology changed into compact colonies in a 2i medium (containing two inhibitors for GSK3 and MEK/ERK). Expression levels of self-renewal marker genes, including Oct3/4 and Nanog, were similar between wild-type and dKO ES cells, whereas proliferation of Etv4/5 dKO ES cells was decreased with overexpression of cyclin-dependent kinase inhibitors (p16/p19, p15, and p57). A differentiation assay revealed that the embryoid bodies derived from Etv4/5 dKO ES cells were smaller than the control, and expression of ectoderm marker genes, including Fgf5, Sox1, and Pax3, was not induced in dKO-derived embryoid bodies. Microarray analysis demonstrated that stem cell-related genes, including Tcf15, Gbx2, Lrh1, Zic3, and Baf60c, were significantly repressed in Etv4/5 dKO ES cells. The artificial expression of Etv4 and/or Etv5 in Etv4/5 dKO ES cells induced re-expression of Tcf15 and Gbx2. These results indicate that Etv4 and Etv5, potentially through regulation of Gbx2 and Tcf15, are involved in the ES cell proliferation and induction of differentiation-associated genes in ES cells.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , DNA-Binding Proteins/genetics , Embryonic Stem Cells/cytology , Glycogen Synthase Kinase 3/biosynthesis , Glycogen Synthase Kinase 3/genetics , Mice , Mice, Knockout , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics
7.
Hum Mol Genet ; 23(25): 6807-14, 2014 Dec 20.
Article in English | MEDLINE | ID: mdl-25082826

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) affect about 1 in 500 births and are a major cause of morbidity in infants. Duplex collecting systems rank among the most common abnormalities of CAKUT, but the molecular basis for this defect is poorly understood. In mice, conditional deletion of Wnt5a in mesoderm results in bilateral duplex kidney and ureter formation. The ureteric buds (UBs) in mutants emerge as doublets from the intermediate mesoderm (IM)-derived nephric duct (ND) without anterior expansion of the glial cell line-derived neurotrophic factor (Gdnf) expression domain in the surrounding mesenchyme. Wnt5a is normally expressed in a graded manner at the posterior end of the IM, but its expression is down-regulated prior to UB outgrowth at E10.5. Furthermore, ablation of Wnt5a in the mesoderm with an inducible Cre at E7.5 results in duplex UBs, whereas ablation at E8.5 yields normal UB outgrowth, demonstrating that Wnt5a functions in IM development well before the formation of the metanephros. In mutants, the posterior ND is duplicated and surrounding Pax2-positive mesenchymal cells persist in the nephric cord, suggesting that disruption of normal ND patterning prompts the formation of duplex ureters and kidneys. Ror2 homozygous mutants, which infrequently yield duplex collecting systems, show a dramatic increase in incidence with the additional deletion of one copy of Wnt5a, implicating this receptor in non-canonical Wnt5a signaling during IM development. This work provides the first evidence of a role of Wnt5a/Ror2 signaling in IM extension and offers new insights into the etiology of CAKUT and possible involvement of Wnt5a/Ror2 mutations.


Subject(s)
Kidney/metabolism , Mesoderm/metabolism , Morphogenesis/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Signal Transduction/genetics , Wnt Proteins/genetics , Animals , Embryo, Mammalian , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Homozygote , Integrases/genetics , Integrases/metabolism , Kidney/growth & development , Kidney/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mesoderm/growth & development , Mesoderm/pathology , Mice , Mice, Transgenic , PAX2 Transcription Factor/genetics , PAX2 Transcription Factor/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Time Factors , Ureter/growth & development , Ureter/metabolism , Ureter/pathology , Wnt Proteins/deficiency , Wnt-5a Protein , Wolffian Ducts/growth & development , Wolffian Ducts/metabolism , Wolffian Ducts/pathology
8.
Development ; 141(17): 3420-30, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25139858

ABSTRACT

The vesico-ureteric junction (VUJ) forms through a complex developmental program that connects the primordium of the upper urinary tract [the nephric duct (ND)] with that of the lower urinary tract (the cloaca). The signals that orchestrate the various tissue interactions in this program are poorly understood. Here, we show that two members of the EphA subfamily of receptor tyrosine kinases, EphA4 and EphA7, are specifically expressed in the mesenchyme surrounding the caudal ND and the cloaca, and that Epha4(-/-);Epha7(+/-) and Epha4(-/-);Epha7(-/-) (DKO) mice display distal ureter malformations including ureterocele, blind and ectopically ending ureters with associated hydroureter, megaureter and hydronephrosis. We trace these defects to a late or absent fusion of the ND with the cloaca. In DKO embryos, the ND extends normally and approaches the cloaca but the tip subsequently looses its integrity. Expression of Gata3 and Lhx1 and their downstream target Ret is severely reduced in the caudal ND. Conditional deletion of ephrin B2 from the ND largely phenocopies these changes, suggesting that EphA4/EphA7 from the pericloacal mesenchyme signal via ephrin B2 to mediate ND insertion. Disturbed activity of this signaling module may entail defects of the VUJ, which are frequent in the spectrum of congenital anomalies of the kidney and the urinary tract (CAKUT) in human newborns.


Subject(s)
Cloaca/embryology , Mesoderm/embryology , Nephrons/embryology , Nephrons/metabolism , Receptor, EphA4/metabolism , Receptor, EphA7/metabolism , Signal Transduction , Animals , Cloaca/metabolism , Cloaca/pathology , Disease Progression , Down-Regulation , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Ephrin-B2/metabolism , GATA3 Transcription Factor/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Humans , Hydronephrosis/embryology , Hydronephrosis/genetics , Hydronephrosis/pathology , Kidney/abnormalities , Kidney/enzymology , Kidney/metabolism , Kidney/pathology , LIM-Homeodomain Proteins/metabolism , Membrane Fusion , Mesoderm/metabolism , Mesoderm/pathology , Mice , Mice, Knockout , Nephrons/pathology , PAX2 Transcription Factor/metabolism , Phenotype , Proto-Oncogene Proteins c-ret/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Ureter/abnormalities , Ureter/embryology , Ureter/metabolism , Ureter/pathology
9.
PLoS Genet ; 10(3): e1004193, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24603431

ABSTRACT

Although the growth factor (GF) signaling guiding renal branching is well characterized, the intracellular cascades mediating GF functions are poorly understood. We studied mitogen-activated protein kinase (MAPK) pathway specifically in the branching epithelia of developing kidney by genetically abrogating the pathway activity in mice lacking simultaneously dual-specificity protein kinases Mek1 and Mek2. Our data show that MAPK pathway is heterogeneously activated in the subset of G1- and S-phase epithelial cells, and its tissue-specific deletion results in severe renal hypodysplasia. Consequently to the deletion of Mek1/2, the activation of ERK1/2 in the epithelium is lost and normal branching pattern in mutant kidneys is substituted with elongation-only phenotype, in which the epithelium is largely unable to form novel branches and complex three-dimensional patterns, but able to grow without primary defects in mitosis. Cellular characterization of double mutant epithelium showed increased E-cadherin at the cell surfaces with its particular accumulation at baso-lateral locations. This indicates changes in cellular adhesion, which were revealed by electron microscopic analysis demonstrating intercellular gaps and increased extracellular space in double mutant epithelium. When challenged to form monolayer cultures, the mutant epithelial cells were impaired in spreading and displayed strong focal adhesions in addition to spiky E-cadherin. Inhibition of MAPK activity reduced paxillin phosphorylation on serine 83 while remnants of phospho-paxillin, together with another focal adhesion (FA) protein vinculin, were augmented at cell surface contacts. We show that MAPK activity is required for branching morphogenesis, and propose that it promotes cell cycle progression and higher cellular motility through remodeling of cellular adhesions.


Subject(s)
Focal Adhesions/genetics , Kidney/growth & development , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Animals , Epithelial Cells/metabolism , Kidney/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinases/genetics , Morphogenesis/genetics , Phosphorylation , Signal Transduction/genetics , Vinculin/metabolism
10.
Cell Rep ; 7(1): 127-37, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24656820

ABSTRACT

Nephrons, the functional units of the kidney, develop from progenitor cells (cap mesenchyme [CM]) surrounding the epithelial ureteric bud (UB) tips. Reciprocal signaling between UB and CM induces nephrogenesis and UB branching. Although low nephron number is implicated in hypertension and renal disease, the mechanisms that determine nephron number are obscure. To test the importance of nephron progenitor cell number, we genetically ablated 40% of these cells, asking whether this would limit kidney size and nephron number or whether compensatory mechanisms would allow the developing organ to recover. The reduction in CM cell number decreased the rate of branching, which in turn allowed the number of CM cells per UB tip to normalize, revealing a self-correction mechanism. However, the retarded UB branching impaired kidney growth, leaving a permanent nephron deficit. Thus, the number of fetal nephron progenitor cells is an important determinant of nephron endowment, largely via its effect on UB branching.


Subject(s)
Kidney/cytology , Kidney/embryology , Nephrons/embryology , Stem Cells/cytology , Animals , Female , Mice , Morphogenesis , Nephrons/cytology , Pregnancy
11.
Methods Mol Biol ; 1075: 305-20, 2014.
Article in English | MEDLINE | ID: mdl-24052360

ABSTRACT

The structure of the ureteric tree in developing mouse and rat kidneys has previously been quantified in two dimensions. While this type of analysis may provide evidence of changes in ureteric growth, these measurements are effectively inaccurate, as the ureteric tree is a three-dimensional (3D) object. Here we describe a method for measuring the ureteric tree in three dimensions. This technique involves (1) culture of the metanephric kidney at embryonic day 12 (mouse) or 14 (rat), (2) whole-mount immunofluorescence to selectively stain ureteric tree epithelium, (3) confocal microscopy to obtain a complete Z series through the ureteric tree, and (4) image analysis algorithms to binarize, skeletonize, and measure individual branch lengths in 3D. This method has been extended to analysis of the same ureteric tree over time (4D). The results obtained provide accurate and precise quantitation of ureteric tree growth in the developing mouse or rat kidney.


Subject(s)
Kidney/growth & development , Microscopy, Confocal/methods , Morphogenesis , Animals , Epithelium/ultrastructure , Imaging, Three-Dimensional , Kidney/ultrastructure , Mice , Organ Culture Techniques , Rats , Ureter/embryology , Ureter/ultrastructure
12.
Dev Cell ; 27(3): 319-30, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24183650

ABSTRACT

The ureteric bud is an epithelial tube that undergoes branching morphogenesis to form the renal collecting system. Although development of a normal kidney depends on proper ureteric bud morphogenesis, the cellular events underlying this process remain obscure. Here, we used time-lapse microscopy together with several genetic labeling methods to observe ureteric bud cell behaviors in developing mouse kidneys. We observed an unexpected cell behavior in the branching tips of the ureteric bud, which we term "mitosis-associated cell dispersal." Premitotic ureteric tip cells delaminate from the epithelium and divide within the lumen; although one daughter cell retains a basal process, allowing it to reinsert into the epithelium at the site of origin, the other daughter cell reinserts at a position one to three cell diameters away. Given the high rate of cell division in ureteric tips, this cellular behavior causes extensive epithelial cell rearrangements that may contribute to renal branching morphogenesis.


Subject(s)
Epithelial Cells/cytology , Homeodomain Proteins/physiology , Kidney/cytology , Mitosis/physiology , Morphogenesis , Ureter/cytology , Animals , Cell Movement , Epithelial Cells/metabolism , Fluorescent Antibody Technique , Kidney/metabolism , Mice , Mice, Knockout , Ureter/metabolism
13.
Wiley Interdiscip Rev Dev Biol ; 1(5): 693-713, 2012.
Article in English | MEDLINE | ID: mdl-22942910

ABSTRACT

The mammalian kidney, which at maturity contains thousands of nephrons joined to a highly branched collecting duct (CD) system, is an important model system for studying the development of a complex organ. Furthermore, congenital anomalies of the kidney and urinary tract, often resulting from defects in ureteric bud branching morphogenesis, are relatively common human birth defects. Kidney development is initiated by interactions between the nephric duct and the metanephric mesenchyme, leading to the outgrowth and repeated branching of the ureteric bud epithelium, which gives rise to the entire renal CD system. Meanwhile, signals from the ureteric bud induce the mesenchyme cells to form the nephron epithelia. This review focuses on development of the CD system, with emphasis on the mouse as an experimental system. The major topics covered include the origin and development of the nephric duct, formation of the ureteric bud, branching morphogenesis of the ureteric bud, and elongation of the CDs. The signals, receptors, transcription factors, and other regulatory molecules implicated in these processes are discussed. In addition, our current knowledge of cellular behaviors that are controlled by these genes and underlie development of the collecting system is reviewed.


Subject(s)
Kidney/growth & development , Nephrons/growth & development , Organogenesis , Ureter/growth & development , Animals , Epithelium/growth & development , Epithelium/metabolism , Gene Expression Regulation, Developmental , Humans , Kidney/metabolism , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Nephrons/metabolism , Ureter/metabolism , Urinary Tract/growth & development , Urinary Tract/metabolism
14.
Development ; 138(24): 5369-78, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22110055

ABSTRACT

During development of the urogenital tract, fibroblast growth factor 8 (Fgf8) is expressed in mesonephric tubules, but its role in this tissue remains undefined. An evaluation of previously generated T-Cre-mediated Fgf8-deficient mice (T-Cre; Fgf8(flox/Δ2,3) mice), which lack Fgf8 expression in the mesoderm, revealed that the cranial region of the Wolffian duct degenerated prematurely and the cranial mesonephric tubules were missing. As a result, the epididymis, vas deferens and efferent ductules were largely absent in mutant mice. Rarb2-Cre was used to eliminate FGF8 from the mesonephric tubules but to allow expression in the adjacent somites. These mutants retained the cranial end of the Wolffian duct and formed the epididymis and vas deferens, but failed to elaborate the efferent ductules, indicating that Fgf8 expression by the mesonephric tubules is required specifically for the formation of the ductules. Ret knockout mice do not form the ureteric bud, a caudal outgrowth of the Wolffian duct and progenitor for the collecting duct network in the kidney, but they do develop the cranial end normally. This indicates that Fgf8, but not Ret, expression is essential to the outgrowth of the cranial mesonephric tubules from the Wolffian duct and to the development of major portions of the sex accessory tissues in the male reproductive tract. Mechanistically, FGF8 functions upstream of Lhx1 expression in forming the nephron, and analysis of Fgf8 mutants similarly shows deficient Lhx1 expression in the mesonephric tubules. These results demonstrate a multifocal requirement for FGF8 in establishing the male reproductive tract ducts and implicate Lhx1 signaling in tubule elongation.


Subject(s)
Fibroblast Growth Factor 8/metabolism , Genitalia, Male/growth & development , Wolffian Ducts/growth & development , Animals , Gene Expression Regulation, Developmental , Genitalia, Male/metabolism , LIM-Homeodomain Proteins/metabolism , Male , Mesoderm/growth & development , Mesoderm/metabolism , Mice , Mice, Knockout , Nephrons/growth & development , Nephrons/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Transcription Factors/metabolism , Urogenital System/growth & development , Urogenital System/metabolism , Wolffian Ducts/metabolism
15.
Dev Biol ; 360(2): 310-7, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22015719

ABSTRACT

The tyrosine phosphatase Shp2 acts downstream of various growth factors, hormones or cytokine receptors. Mutations of the Shp2 gene are associated with several human diseases. Here we have ablated Shp2 in the developing kidneys of mice, using the ureteric bud epithelium-specific Hoxb7/Cre. Mutant mice produced a phenotype that is similar to mutations of the genes of the GDNF/Ret receptor system, that is: strongly reduced ureteric bud branching and downregulation of the Ret target genes Etv4 and Etv5. Shp2 mutant embryonic kidneys also displayed reduced cell proliferation at the branch tips and branching defects, which could not be overcome by GDNF in organ culture. We also examined compound mutants of Shp2 and Sprouty1, which is an inhibitor of receptor tyrosine kinase signaling in the kidney. Sprouty1 single mutants produce supernumerary ureteric buds, which branch excessively. Sprouty1 mutants rescued branching deficits in Ret(-/-) and GDNF(-/-) kidneys. Sprouty1; Shp2 double mutants showed no rescue of kidney branching. Our data thus indicate an intricate interplay of Shp2 and Sprouty1 in signaling downstream of receptor tyrosine kinases during kidney development. Apparently, Shp2 mediates not only GDNF/Ret but also signaling by other receptor tyrosine kinases in branching morphogenesis of the embryonic kidney.


Subject(s)
DNA-Binding Proteins/genetics , Glial Cell Line-Derived Neurotrophic Factor/genetics , Kidney/embryology , Nuclear Proteins/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Proliferation , DNA-Binding Proteins/metabolism , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Homeodomain Proteins/genetics , Kidney/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Morphogenesis , Mutation , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction/genetics , Ubiquitin-Protein Ligases
16.
Cold Spring Harb Protoc ; 2011(5): pdb.prot5613, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21536763

ABSTRACT

INTRODUCTION: In this article, we outline procedures for the dissection of intact kidneys and the isolation of the ureteric bud (UB) and the metanephric mesenchyme (MM) from mouse embryos. The apparatus required for the culture of these tissues in vitro is described in detail as well as the equipment necessary for performing time-lapse imaging studies of the developing kidney.


Subject(s)
Dissection/methods , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/cytology , Kidney/embryology , Time-Lapse Imaging/methods , Animals , Kidney/anatomy & histology , Kidney/cytology , Mice , Morphogenesis , Ureter/anatomy & histology , Ureter/cytology , Ureter/embryology
17.
Cold Spring Harb Protoc ; 2011(5): pdb.top109, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21536773

ABSTRACT

INTRODUCTION: Development of the kidney involves interactions between several cell lineages and complex morphogenetic processes, such as branching of the ureteric bud (UB) to form the collecting duct system and condensation and differentiation of the mesenchymal progenitors to form the nephron epithelia. One of the advantages of the mouse kidney as an experimental system is that it can develop in culture, from the stage of initial branching of the UB (E11.5) for up to a week (although it achieves the size and degree of development of only an E13.5-E14.5 kidney in vivo). The availability of fluorescent proteins (FPs) has provided powerful tools for visualizing the morphogenesis of specific renal structures in organ cultures. Two categories of genetically modified mice that express FPs are useful for visualizing different cell lineages and developmental processes in these organ cultures: (1) transgenic mice that express a fluorescent reporter in the pattern of a specific gene; and (2) Cre reporter mice, which turn on an FP in cells with Cre recombinase activity (and their daughter cells), used in conjunction with cell type-specific Cre transgenic mice. Here, we describe some of the currently available Cre and FP transgenic lines that are useful for the study of kidney development.


Subject(s)
Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/cytology , Kidney/embryology , Animals , Genes, Reporter , Integrases/genetics , Integrases/metabolism , Kidney/anatomy & histology , Kidney/cytology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Transgenic , Morphogenesis , Ureter/anatomy & histology , Ureter/cytology , Ureter/embryology
18.
Hum Mol Genet ; 20(6): 1143-53, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21212101

ABSTRACT

Congenital abnormalities of the kidney and urinary tract are some of the most common defects detected in the unborn child. Kidney growth is controlled by the GDNF/RET signalling pathway, but the molecular events required for the activation of RET downstream targets are still poorly understood. Here we show that SOX9, a gene involved in campomelic dysplasia (CD) in humans, together with its close homologue SOX8, plays an essential role in RET signalling. Expression of SOX9 can be found from the earliest stages of renal development within the ureteric tip, the ureter mesenchyme and in a segment-specific manner during nephrogenesis. Using a tissue-specific knockout approach, we show that, in the ureteric tip, SOX8 and SOX9 are required for ureter branching, and double-knockout mutants exhibit severe kidney defects ranging from hypoplastic kidneys to renal agenesis. Further genetic analysis shows that SOX8/9 are required downstream of GDNF signalling for the activation of RET effector genes such as Sprouty1 and Etv5. At later stages of development, SOX9 is required to maintain ureteric tip identity and SOX9 ablation induces ectopic nephron formation. Taken together, our study shows that SOX9 acts at multiple steps during kidney organogenesis and identifies SOX8 and SOX9 as key factors within the RET signalling pathway. Our results also explain the aetiology of kidney hypoplasia found in a proportion of CD patients.


Subject(s)
Campomelic Dysplasia/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Kidney/embryology , Proto-Oncogene Proteins c-ret/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction , Animals , Campomelic Dysplasia/embryology , Campomelic Dysplasia/genetics , Cells, Cultured , Disease Models, Animal , Female , Humans , Kidney/metabolism , Male , Mice , Mice, Knockout , Organogenesis , Proto-Oncogene Proteins c-ret/genetics , SOX9 Transcription Factor/genetics , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism
19.
Neurobiol Aging ; 32(12): 2316.e1-15, 2011 Dec.
Article in English | MEDLINE | ID: mdl-20409609

ABSTRACT

The possibility that the degeneration of hippocampal neurons can be caused by mis-regulation of Wnt/ß-catenin signaling was tested. Downregulation of Wnt signaling by the inducible expression of Axin, ICAT, and dnTcf4E causes degeneration of hippocampal neurons, while upregulation of Wnt signaling by the inducible expression of Dvl and ß-catenin has a negligible effect. Treatment with ICG-001, a small molecule known to inhibit Wnt signaling, causes degeneration of hippocampal neurons, while the treatment with a JNK specific inhibitor does not show any effect. The results from LDH and TUNEL assays suggest that degeneration occurs via apoptotic processes. Inhibition of Wnt signaling reduced IGF-1 expression and the addition of IGF-1 blocked degeneration, which suggests that downregulation of IGF-1/Akt signaling is partially responsible for the degeneration. Inducible expression of Axin in the hippocampal neurons isolated from Axin2P-rtTA/pBI-EGFP-Axin double transgenic mice also causes degeneration. Consistent with the findings, these mice had more neuronal cell death in hippocampus and had differences in contextual conditioning upon the inducible expression of Axin. In summary, our data strongly support the idea that downregulation of Wnt/ß-catenin signaling causes degeneration of hippocampal neurons in vivo and may be a cause of neurodegenerative diseases related to an anxiety related response.


Subject(s)
Hippocampus/pathology , Nerve Degeneration/pathology , Neurons/physiology , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , beta Catenin/antagonists & inhibitors , Animals , Anxiety/psychology , Down-Regulation/physiology , Hippocampus/physiology , Mice , Mice, Inbred ICR , Mice, Transgenic , Nerve Degeneration/metabolism , Neurons/pathology , Wnt Proteins/antagonists & inhibitors , beta Catenin/physiology
20.
PLoS Genet ; 6(10): e1001176, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-21060807

ABSTRACT

The actin depolymerizing factors (ADFs) play important roles in several cellular processes that require cytoskeletal rearrangements, such as cell migration, but little is known about the in vivo functions of ADFs in developmental events like branching morphogenesis. While the molecular control of ureteric bud (UB) branching during kidney development has been extensively studied, the detailed cellular events underlying this process remain poorly understood. To gain insight into the role of actin cytoskeletal dynamics during renal branching morphogenesis, we studied the functional requirements for the closely related ADFs cofilin1 (Cfl1) and destrin (Dstn) during mouse development. Either deletion of Cfl1 in UB epithelium or an inactivating mutation in Dstn has no effect on renal morphogenesis, but simultaneous lack of both genes arrests branching morphogenesis at an early stage, revealing considerable functional overlap between cofilin1 and destrin. Lack of Cfl1 and Dstn in the UB causes accumulation of filamentous actin, disruption of normal epithelial organization, and defects in cell migration. Animals with less severe combinations of mutant Cfl1 and Dstn alleles, which retain one wild-type Cfl1 or Dstn allele, display abnormalities including ureter duplication, renal hypoplasia, and abnormal kidney shape. The results indicate that ADF activity, provided by either cofilin1 or destrin, is essential in UB epithelial cells for normal growth and branching.


Subject(s)
Cofilin 1/metabolism , Destrin/metabolism , Morphogenesis , Ureter/metabolism , Actins/metabolism , Animals , Cell Movement , Cofilin 1/genetics , Destrin/genetics , Epithelial Cells/metabolism , Epithelium/embryology , Epithelium/metabolism , Female , Fluorescent Antibody Technique , Genotype , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , In Situ Hybridization , Kidney/drug effects , Kidney/embryology , Kidney/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Ureter/drug effects , Ureter/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...