ureteric bud
Recently Published Documents


TOTAL DOCUMENTS

277
(FIVE YEARS 34)

H-INDEX

43
(FIVE YEARS 2)

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Alison E. Jarmas ◽  
Eric W. Brunskill ◽  
Praneet Chaturvedi ◽  
Nathan Salomonis ◽  
Raphael Kopan

AbstractMammalian nephron endowment is determined by the coordinated cessation of nephrogenesis in independent niches. Here we report that translatome analysis in Tsc1+/− nephron progenitor cells from mice with elevated nephron numbers reveals how differential translation of Wnt antagonists over agonists tips the balance between self-renewal and differentiation. Wnt agonists are poorly translated in young niches, resulting in an environment with low R-spondin and high Fgf20 promoting self-renewal. In older niches we find increased translation of Wnt agonists, including R-spondin and the signalosome-promoting Tmem59, and low Fgf20, promoting differentiation. This suggests that the tipping point for nephron progenitor exit from the niche is controlled by the gradual increase in stability and possibly clustering of Wnt/Fzd complexes in individual cells, enhancing the response to ureteric bud-derived Wnt9b inputs and driving synchronized differentiation. As predicted by these findings, removing one Rspo3 allele in nephron progenitors delays cessation and increases nephron numbers in vivo.


2021 ◽  
Vol 18 ◽  
pp. 170-182
Author(s):  
Afshin Poorkhanalikoudehi ◽  
Karl-Heinz Zimmermann

Epithelium is a complex component in the mammalian kidney that has a highly branched duct system. Branching morphogenesis has a hierarchy structure in the ureteric bud and produces the collecting duct tree through repetitive processes. Epithelial and mesenchymal cells surround the tips of growing branches, and their cellular reactions adjust the ureteric bud branching. Mesenchymal cells produce a small protein called glial cellline derived neurotrophic factor (GDNF) that connects to te Rearranged in Transfection (RET) receptors on the surface of epithelial cells. The identified reactions are a necessity for the normal branching growth and their roles exist for using biological features in the proposed model. This paper presents an agent-based model based on cellular automaton for kidney branching in ex-vivo using the features that are expressed as artificial patterns in algorithms. This model extending the groundbreaking approach of Lambert et al. is flexible in features and high compatibility with experimental data. Mesenchymal cells and RET receptors are also expressed as mathematical patterns in the algorithms. The growth mechanism is determined by the growth factor, which indicates the epithelial cell branch when its cell division depends on the local concentration growth factor. Cell division occurs when the level of stimulus growth factor exceeds the threshold. Comparison shows that the model mimics experimental data with high consistency and reveals the dependence between growth factor parameters and features. Results indicate the superiority of compatibility with nature when compared with the model mentioned above.


2021 ◽  
Vol 220 (11) ◽  
Author(s):  
Fabian Bock ◽  
Bertha C. Elias ◽  
Xinyu Dong ◽  
Diptiben V. Parekh ◽  
Glenda Mernaugh ◽  
...  

A polarized collecting duct (CD), formed from the branching ureteric bud (UB), is a prerequisite for an intact kidney. The small Rho GTPase Rac1 is critical for actin cytoskeletal regulation. We investigated the role of Rac1 in the kidney collecting system by selectively deleting it in mice at the initiation of UB development. The mice exhibited only a mild developmental phenotype; however, with aging, the CD developed a disruption of epithelial integrity and function. Despite intact integrin signaling, Rac1-null CD cells had profound adhesion and polarity abnormalities that were independent of the major downstream Rac1 effector, Pak1. These cells did however have a defect in the WAVE2–Arp2/3 actin nucleation and polymerization apparatus, resulting in actomyosin hyperactivity. The epithelial defects were reversible with direct myosin II inhibition. Furthermore, Rac1 controlled lateral membrane height and overall epithelial morphology by maintaining lateral F-actin and restricting actomyosin. Thus, Rac1 promotes CD epithelial integrity and morphology by restricting actomyosin via Arp2/3-dependent cytoskeletal branching.


2021 ◽  
Author(s):  
Michael A Ortega ◽  
Ross K Villiger ◽  
Malia Harrison-Chau ◽  
Suzanna Lieu ◽  
Kadee-Kalia Tamashiro ◽  
...  

Ureter obstruction is a highly prevalent event during embryonic development and is a major cause of pediatric kidney disease. We have reported that ureteric bud specific ablation of the exocyst Exoc5 subunit in late murine gestation results in failure of urothelial stratification, cell death, and complete ureter obstruction. However, the mechanistic connection between disrupted exocyst activity, urothelial cell death, and subsequent ureter obstruction was unclear. Here, we report that inhibited urothelial stratification does not drive cell death during ureter development. Instead, we demonstrate that the exocyst plays a critical role in autophagy in urothelial cells, and that disruption of autophagy activates a urothelial NF-κB stress response. Impaired autophagy first provokes canonical NF κB activity which is progressively followed by increasing non-canonical NF-κB activity and cell death if the stress remains unresolved. Furthermore, we demonstrate that ureter obstructions can be completely rescued in Exoc5 conditional knockout mice by administering a single dose of pan-caspase inhibitor z VAD-FMK at E16.5 prior to urothelial cell death. Taken together, ablation of Exoc5 disrupts autophagic stress response and activates progressive NF-κB signaling which promotes obstructive uropathy.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Zipeng Zeng ◽  
Biao Huang ◽  
Riana K. Parvez ◽  
Yidan Li ◽  
Jyunhao Chen ◽  
...  

AbstractCurrent kidney organoids model development and diseases of the nephron but not the contiguous epithelial network of the kidney’s collecting duct (CD) system. Here, we report the generation of an expandable, 3D branching ureteric bud (UB) organoid culture model that can be derived from primary UB progenitors from mouse and human fetal kidneys, or generated de novo from human pluripotent stem cells. In chemically-defined culture conditions, UB organoids generate CD organoids, with differentiated principal and intercalated cells adopting spatial assemblies reflective of the adult kidney’s collecting system. Aggregating 3D-cultured nephron progenitor cells with UB organoids in vitro results in a reiterative process of branching morphogenesis and nephron induction, similar to kidney development. Applying an efficient gene editing strategy to remove RET activity, we demonstrate genetically modified UB organoids can model congenital anomalies of kidney and urinary tract. Taken together, these platforms will facilitate an enhanced understanding of development, regeneration and diseases of the mammalian collecting duct system.


Development ◽  
2021 ◽  
Vol 148 (10) ◽  
Author(s):  
Hao Li ◽  
Kristen Kurtzeborn ◽  
Jussi Kupari ◽  
Yujuan Gui ◽  
Edward Siefker ◽  
...  

ABSTRACT Nephron endowment, defined during the fetal period, dictates renal and related cardiovascular health throughout life. We show here that, despite its negative effects on kidney growth, genetic increase of GDNF prolongs the nephrogenic program beyond its normal cessation. Multi-stage mechanistic analysis revealed that excess GDNF maintains nephron progenitors and nephrogenesis through increased expression of its secreted targets and augmented WNT signaling, leading to a two-part effect on nephron progenitor maintenance. Abnormally high GDNF in embryonic kidneys upregulates its known targets but also Wnt9b and Axin2, with concomitant deceleration of nephron progenitor proliferation. Decline of GDNF levels in postnatal kidneys normalizes the ureteric bud and creates a permissive environment for continuation of the nephrogenic program, as demonstrated by morphologically and molecularly normal postnatal nephron progenitor self-renewal and differentiation. These results establish that excess GDNF has a bi-phasic effect on nephron progenitors in mice, which can faithfully respond to GDNF dosage manipulation during the fetal and postnatal period. Our results suggest that sensing the signaling activity level is an important mechanism through which GDNF and other molecules contribute to nephron progenitor lifespan specification.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Oraly Sanchez-Ferras ◽  
Alain Pacis ◽  
Maria Sotiropoulou ◽  
Yuhong Zhang ◽  
Yu Chang Wang ◽  
...  

AbstractThe kidney and upper urinary tract develop through reciprocal interactions between the ureteric bud and the surrounding mesenchyme. Ureteric bud branching forms the arborized collecting duct system of the kidney, while ureteric tips promote nephron formation from dedicated progenitor cells. While nephron progenitor cells are relatively well characterized, the origin of ureteric bud progenitors has received little attention so far. It is well established that the ureteric bud is induced from the nephric duct, an epithelial duct derived from the intermediate mesoderm of the embryo. However, the cell state transitions underlying the progression from intermediate mesoderm to nephric duct and ureteric bud remain unknown. Here we show that nephric duct morphogenesis results from the coordinated organization of four major progenitor cell populations. Using single cell RNA-seq and Cluster RNA-seq, we show that these progenitors emerge in time and space according to a stereotypical pattern. We identify the transcription factors Tfap2a/b and Gata3 as critical coordinators of this progenitor cell progression. This study provides a better understanding of the cellular origin of the renal collecting duct system and associated urinary tract developmental diseases, which may inform guided differentiation of functional kidney tissue.


Development ◽  
2021 ◽  
Vol 148 (9) ◽  
Author(s):  
Adam Packard ◽  
William H. Klein ◽  
Frank Costantini

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.


2021 ◽  
Vol 8 (1) ◽  
Author(s):  
Stefan Kohl ◽  
Sandra Habbig ◽  
Lutz T. Weber ◽  
Max C. Liebau

AbstractCongenital anomalies of the kidney and urinary tract (CAKUT) occur in 0.5–1/100 newborns and as a group they represent the most frequent cause for chronic kidney failure in children. CAKUT comprise clinically heterogeneous conditions, ranging from mild vesicoureteral reflux to kidney aplasia. Most forms of CAKUT share the pathophysiology of an impaired developmental interaction of the ureteric bud (UB) and the metanephric mesenchyme (MM). In most cases, CAKUT present as an isolated condition. They also may occur as a component in rare multi-organ syndromes. Many CAKUT probably have a multifactorial etiology. However, up to 20% of human patients and > 200 transgenic mouse models have a monogenic form of CAKUT, which has fueled our efforts to unravel molecular kidney (mal-)development. To date, genetic variants in more than 50 genes have been associated with (isolated) CAKUT in humans. In this short review, we will summarize typical imaging findings in patients with CAKUT and highlight recent mechanistic insight in the molecular pathogenesis of monogenic forms of CAKUT.


Sign in / Sign up

Export Citation Format

Share Document