Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38496522

ABSTRACT

The kidney functions as a finely tuned sensor to balance body fluid composition and filter out waste through complex coordinated mechanisms. This versatility requires tight neural control, with innervating efferent nerves playing a crucial role in regulating blood flow, glomerular filtration rate, water and sodium reabsorption, and renin release. In turn sensory afferents provide feedback to the central nervous system for the modulation of cardiovascular function. However, the cells targeted by sensory afferents and the physiological sensing mechanisms remain poorly characterized. Moreover, how the kidney is innervated during development to establish these functions remains elusive. Here, we utilized a combination of light-sheet and confocal microscopy to generate anatomical maps of kidney sensory and sympathetic nerves throughout development and resolve the establishment of functional crosstalk. Our analyses revealed that kidney innervation initiates at embryonic day (E)13.5 as the nerves associate with vascular smooth muscle cells and follow arterial differentiation. By E17.5 axonal projections associate with kidney structures such as glomeruli and tubules and the network continues to expand postnatally. These nerves are synapsin I-positive, highlighting ongoing axonogenesis and the potential for functional crosstalk. We show that sensory and sympathetic nerves innervate the kidney concomitantly and classify the sensory fibers as calcitonin gene related peptide (CGRP)+, substance P+, TRPV1+, and PIEZO2+, establishing the presence of PIEZO2 mechanosensory fibers in the kidney. Using retrograde tracing, we identified the primary dorsal root ganglia, T10-L2, from which PIEZO2+ sensory afferents project to the kidney. Taken together our findings elucidate the temporality of kidney innervation and resolve the identity of kidney sympathetic and sensory nerves.

2.
Development ; 150(22)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37818607

ABSTRACT

The intricate vascular system of the kidneys supports body fluid and organ homeostasis. However, little is known about how vascular architecture is established during kidney development. More specifically, how signals from the kidney influence vessel maturity and patterning remains poorly understood. Netrin 1 (Ntn1) is a secreted ligand that is crucial for vessel and neuronal guidance. Here, we demonstrate that Ntn1 is expressed by Foxd1+ stromal progenitors in the developing mouse kidney and conditional deletion (Foxd1GC/+;Ntn1fl/fl) results in hypoplastic kidneys with extended nephrogenesis. Wholemount 3D analyses additionally revealed the loss of a predictable vascular pattern in Foxd1GC/+;Ntn1fl/fl kidneys. As vascular patterning has been linked to vessel maturity, we investigated arterialization. Quantification of the CD31+ endothelium at E15.5 revealed no differences in metrics such as the number of branches or branch points, whereas the arterial vascular smooth muscle metrics were significantly reduced at both E15.5 and P0. In support of our observed phenotypes, whole kidney RNA-seq revealed disruptions to genes and programs associated with stromal cells, vasculature and differentiating nephrons. Together, our findings highlight the significance of Ntn1 to proper vascularization and kidney development.


Subject(s)
Kidney , Nephrons , Animals , Mice , Netrin-1/genetics , Phenotype
3.
Dis Model Mech ; 16(11)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37815464

ABSTRACT

Wilms tumors present as an amalgam of varying proportions of tissues located within the developing kidney, one being the nephrogenic blastema comprising multipotent nephron progenitor cells (NPCs). The recurring missense mutation Q177R in NPC transcription factors SIX1 and SIX2 is most correlated with tumors of blastemal histology and is significantly associated with relapse. Yet, the transcriptional regulatory consequences of SIX1/2-Q177R that might promote tumor progression and recurrence have not been investigated extensively. Utilizing multiple Wilms tumor transcriptomic datasets, we identified upregulation of the gene encoding non-canonical WNT ligand WNT5A in addition to other WNT pathway effectors in SIX1/2-Q177R mutant tumors. SIX1 ChIP-seq datasets from Wilms tumors revealed shared binding sites for SIX1/SIX1-Q177R within a promoter of WNT5A and at putative distal cis-regulatory elements (CREs). We demonstrate colocalization of SIX1 and WNT5A in Wilms tumor tissue and utilize in vitro assays that support SIX1 and SIX1-Q177R activation of expression from the WNT5A CREs, as well as enhanced binding affinity within the WNT5A promoter that may promote the differential expression of WNT5A and other WNT pathway effectors associated with SIX1-Q177R tumors.


Subject(s)
Kidney Neoplasms , Wilms Tumor , Humans , Wnt Signaling Pathway , Gene Expression Regulation, Neoplastic , Neoplasm Recurrence, Local/genetics , Wilms Tumor/genetics , Wilms Tumor/metabolism , Wilms Tumor/pathology , Wnt-5a Protein/genetics , Wnt-5a Protein/metabolism , Kidney Neoplasms/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
4.
Front Cell Dev Biol ; 11: 1195037, 2023.
Article in English | MEDLINE | ID: mdl-37325559

ABSTRACT

Introduction: The unique architecture of glomerular podocytes is integral to kidney filtration. Interdigitating foot processes extend from the podocyte cell body, wrap around fenestrated capillaries, and form specialized junctional complexes termed slit diaphragms to create a molecular sieve. However, the full complement of proteins which maintain foot process integrity, and how this localized proteome changes with disease, remain to be elucidated. Methods: Proximity-dependent biotin identification (BioID) enables the identification of spatially localized proteomes. To this end, we developed a novel in vivo BioID knock-in mouse model. We utilized the slit diaphragm protein podocin (Nphs2) to create a podocin-BioID fusion. Podocin-BioID localizes to the slit diaphragm, and biotin injection leads to podocyte-specific protein biotinylation. We isolated the biotinylated proteins and performed mass spectrometry to identify proximal interactors. Results and Discussion: Gene ontology analysis of 54 proteins specifically enriched in our podocin-BioID sample revealed 'cell junctions,' 'actin binding,' and 'cytoskeleton organization' as top terms. Known foot process components were identified, and we further uncovered two novel proteins: the tricellular junctional protein Ildr2 and the CDC42 and N-WASP interactor Fnbp1l. We confirmed that Ildr2 and Fnbp1l are expressed by podocytes and partially colocalize with podocin. Finally, we investigated how this proteome changes with age and uncovered a significant increase in Ildr2. This was confirmed by immunofluorescence on human kidney samples and suggests altered junctional composition may preserve podocyte integrity. Together, these assays have led to new insights into podocyte biology and support the efficacy of utilizing BioID in vivo to interrogate spatially localized proteomes in health, aging, and disease.

5.
bioRxiv ; 2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37131589

ABSTRACT

Blood filtering by the kidney requires the establishment of an intricate vascular system that works to support body fluid and organ homeostasis. Despite these critical roles, little is known about how vascular architecture is established during kidney development. More specifically, how signals from the kidney influence vessel maturity and patterning remains poorly understood. Netrin-1 (Ntn1) is a secreted ligand critical for vessel and neuronal guidance. Here, we demonstrate that Ntn1 is expressed by stromal progenitors in the developing kidney, and conditional deletion of Ntn1 from Foxd1+ stromal progenitors (Foxd1GC/+;Ntn1fl/fl) results in hypoplastic kidneys that display extended nephrogenesis. Despite expression of the netrin-1 receptor Unc5c in the adjacent nephron progenitor niche, Unc5c knockout kidneys develop normally. The netrin-1 receptor Unc5b is expressed by embryonic kidney endothelium and therefore we interrogated the vascular networks of Foxd1GC/+;Ntn1fl/fl kidneys. Wholemount, 3D analyses revealed the loss of a predictable vascular pattern in mutant kidneys. As vascular patterning has been linked to vessel maturity, we investigated arterialization in these mutants. Quantification of the CD31+ endothelium at E15.5 revealed no differences in metrics such as the number of branches or branch points, whereas the arterial vascular smooth muscle metrics were significantly reduced at both E15.5 and P0. In support of these results, whole kidney RNA-seq showed upregulation of angiogenic programs and downregulation of muscle-related programs which included smooth muscle-associated genes. Together, our findings highlight the significance of netrin-1 to proper vascularization and kidney development.

6.
Cell Rep ; 41(6): 111610, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36351395

ABSTRACT

In both humans and mice, repair of acute kidney injury is worse in males than in females. Here, we provide evidence that this sexual dimorphism results from sex differences in ferroptosis, an iron-dependent, lipid-peroxidation-driven regulated cell death. Using genetic and single-cell transcriptomic approaches in mice, we report that female sex confers striking protection against ferroptosis, which was experimentally induced in proximal tubular (PT) cells by deleting glutathione peroxidase 4 (Gpx4). Single-cell transcriptomic analyses further identify the NFE2-related factor 2 (NRF2) antioxidant protective pathway as a female resilience mechanism against ferroptosis. Genetic inhibition and pharmacological activation studies show that NRF2 controls PT cell fate and plasticity by regulating ferroptosis. Importantly, pharmacological NRF2 activation protects male PT cells from ferroptosis and improves cellular plasticity as in females. Our data highlight NRF2 as a potential therapeutic target to prevent failed renal repair after acute kidney injury in both sexes by modulating cellular plasticity.


Subject(s)
Acute Kidney Injury , Ferroptosis , Humans , Female , Male , Mice , Animals , Sex Characteristics , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Kidney/metabolism
7.
Curr Top Dev Biol ; 148: 195-235, 2022.
Article in English | MEDLINE | ID: mdl-35461566

ABSTRACT

Proper innervation of peripheral organs helps to maintain physiological homeostasis and elicit responses to external stimuli. Disruptions to normal function can result in pathophysiological consequences. The establishment of connections and communication between the central nervous system and the peripheral organs is accomplished through the peripheral nervous system. Neuronal connections with target tissues arise from ganglia partitioned throughout the body. Organ innervation is initiated during development with stimuli being conducted through several types of neurons including sympathetic, parasympathetic, and sensory. While the physiological modulation of mature organs by these nerves is largely understood, their role in mammalian development is only beginning to be uncovered. Interactions with cells in target tissues can affect the development and eventual function of several organs, highlighting their significance. This chapter will cover the origin of peripheral neurons, factors mediating organ innervation, and the composition and function of organ-specific nerves during development. This emerging field aims to identify the functional contribution of innervation to development which will inform future investigations of normal and abnormal mammalian organogenesis, as well as contribute to regenerative and organ replacement efforts where nerve-derived signals may have significant implications for the advancement of such studies.


Subject(s)
Nervous System Physiological Phenomena , Neurons , Animals , Central Nervous System , Mammals , Neurons/physiology , Organogenesis , Peripheral Nervous System
8.
Elife ; 102021 07 19.
Article in English | MEDLINE | ID: mdl-34279220

ABSTRACT

Overwhelming lipid peroxidation induces ferroptotic stress and ferroptosis, a non-apoptotic form of regulated cell death that has been implicated in maladaptive renal repair in mice and humans. Using single-cell transcriptomic and mouse genetic approaches, we show that proximal tubular (PT) cells develop a molecularly distinct, pro-inflammatory state following injury. While these inflammatory PT cells transiently appear after mild injury and return to their original state without inducing fibrosis, after severe injury they accumulate and contribute to persistent inflammation. This transient inflammatory PT state significantly downregulates glutathione metabolism genes, making the cells vulnerable to ferroptotic stress. Genetic induction of high ferroptotic stress in these cells after mild injury leads to the accumulation of the inflammatory PT cells, enhancing inflammation and fibrosis. Our study broadens the roles of ferroptotic stress from being a trigger of regulated cell death to include the promotion and accumulation of proinflammatory cells that underlie maladaptive repair.


Subject(s)
Epithelial Cells/metabolism , Kidney/injuries , Kidney/metabolism , Acute Kidney Injury/genetics , Acute Kidney Injury/therapy , Animals , Cell Death , Ferroptosis/genetics , Fibrosis/genetics , Gene Expression , Inflammation/genetics , Iron/metabolism , Kidney/pathology , Lipid Peroxidation , Male , Mice , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Regenerative Medicine
9.
Biotechniques ; 70(3): 181-185, 2021 03.
Article in English | MEDLINE | ID: mdl-33337254

ABSTRACT

Blood vessels perform critical functions in both health and disease. Understanding how vessels form, pattern and respond to damage is essential. However, labeling and imaging the vasculature to ascertain these properties can be difficult and time-consuming. Here, the authors present a novel methodology for rapidly and efficiently labeling whole vascular networks in vivo by exploiting the fluorescent properties of Evans blue. By combining the labeling with fluorescence microscopy, this method enables visualization of whole tissue vasculature for a fraction of the time and cost compared with traditional methods.


Subject(s)
Blood Vessels/diagnostic imaging , Coloring Agents , Evans Blue , Microscopy, Fluorescence
10.
Genetics ; 216(4): 905-930, 2020 12.
Article in English | MEDLINE | ID: mdl-33067325

ABSTRACT

The laboratory mouse is the most widely used animal model for biomedical research, due in part to its well-annotated genome, wealth of genetic resources, and the ability to precisely manipulate its genome. Despite the importance of genetics for mouse research, genetic quality control (QC) is not standardized, in part due to the lack of cost-effective, informative, and robust platforms. Genotyping arrays are standard tools for mouse research and remain an attractive alternative even in the era of high-throughput whole-genome sequencing. Here, we describe the content and performance of a new iteration of the Mouse Universal Genotyping Array (MUGA), MiniMUGA, an array-based genetic QC platform with over 11,000 probes. In addition to robust discrimination between most classical and wild-derived laboratory strains, MiniMUGA was designed to contain features not available in other platforms: (1) chromosomal sex determination, (2) discrimination between substrains from multiple commercial vendors, (3) diagnostic SNPs for popular laboratory strains, (4) detection of constructs used in genetically engineered mice, and (5) an easy-to-interpret report summarizing these results. In-depth annotation of all probes should facilitate custom analyses by individual researchers. To determine the performance of MiniMUGA, we genotyped 6899 samples from a wide variety of genetic backgrounds. The performance of MiniMUGA compares favorably with three previous iterations of the MUGA family of arrays, both in discrimination capabilities and robustness. We have generated publicly available consensus genotypes for 241 inbred strains including classical, wild-derived, and recombinant inbred lines. Here, we also report the detection of a substantial number of XO and XXY individuals across a variety of sample types, new markers that expand the utility of reduced complexity crosses to genetic backgrounds other than C57BL/6, and the robust detection of 17 genetic constructs. We provide preliminary evidence that the array can be used to identify both partial sex chromosome duplication and mosaicism, and that diagnostic SNPs can be used to determine how long inbred mice have been bred independently from the relevant main stock. We conclude that MiniMUGA is a valuable platform for genetic QC, and an important new tool to increase the rigor and reproducibility of mouse research.


Subject(s)
Genome-Wide Association Study/methods , Genotyping Techniques/methods , Mice/genetics , Oligonucleotide Array Sequence Analysis/methods , Animals , Female , Genome-Wide Association Study/standards , Genotype , Genotyping Techniques/standards , Male , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis/standards , Polymorphism, Genetic , Reproducibility of Results , Sex Determination Processes
11.
Semin Cell Dev Biol ; 91: 94-103, 2019 07.
Article in English | MEDLINE | ID: mdl-30030141

ABSTRACT

The filtering component of the kidney, the nephron, arises from a single progenitor population. These nephron progenitor cells (NPCs) both self-renew and differentiate throughout the course of kidney development ensuring sufficient nephron endowment. An appropriate balance of these processes must be struck as deficiencies in nephron numbers are associated with hypertension and kidney disease. This review will discuss the mechanisms and molecules supporting NPC maintenance and differentiation. A focus on recent work will highlight new molecular insights into NPC regulation and their dynamic behavior in both space and time.


Subject(s)
Kidney/cytology , Nephrons/cytology , Podocytes/cytology , Stem Cells/cytology , Animals , Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Humans , Kidney/embryology , Kidney/metabolism , Nephrons/metabolism , Organogenesis/genetics , Podocytes/metabolism , Stem Cells/metabolism
12.
Elife ; 72018 12 05.
Article in English | MEDLINE | ID: mdl-30516471

ABSTRACT

A normal endowment of nephrons in the mammalian kidney requires a balance of nephron progenitor self-renewal and differentiation throughout development. Here, we provide evidence for a novel action of ureteric branch tip-derived Wnt11 in progenitor cell organization and interactions within the nephrogenic niche, ultimately determining nephron endowment. In Wnt11 mutants, nephron progenitors dispersed from their restricted niche, intermixing with interstitial progenitors. Nephron progenitor differentiation was accelerated, kidneys were significantly smaller, and the nephron progenitor pool was prematurely exhausted, halving the final nephron count. Interestingly, RNA-seq revealed no significant differences in gene expression. Live imaging of nephron progenitors showed that in the absence of Wnt11 they lose stable attachments to the ureteric branch tips, continuously detaching and reattaching. Further, the polarized distribution of several markers within nephron progenitors is disrupted. Together these data highlight the importance of Wnt11 signaling in directing nephron progenitor behavior which determines a normal nephrogenic program.


Subject(s)
Cell Polarity/genetics , Gene Expression Regulation, Developmental , Nephrons/metabolism , Organogenesis/genetics , Stem Cells/metabolism , Wnt Proteins/genetics , Animals , Cell Differentiation , Cell Movement , Embryo, Mammalian , Female , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Keratin-8/genetics , Keratin-8/metabolism , Male , Mice , Mice, Transgenic , Nephrons/cytology , Nephrons/growth & development , Protein Isoforms/genetics , Protein Isoforms/metabolism , Signal Transduction , Stem Cells/cytology , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Proteins/metabolism
13.
PLoS Genet ; 14(1): e1007181, 2018 01.
Article in English | MEDLINE | ID: mdl-29377931

ABSTRACT

Nephron progenitor number determines nephron endowment; a reduced nephron count is linked to the onset of kidney disease. Several transcriptional regulators including Six2, Wt1, Osr1, Sall1, Eya1, Pax2, and Hox11 paralogues are required for specification and/or maintenance of nephron progenitors. However, little is known about the regulatory intersection of these players. Here, we have mapped nephron progenitor-specific transcriptional networks of Six2, Hoxd11, Osr1, and Wt1. We identified 373 multi-factor associated 'regulatory hotspots' around genes closely associated with progenitor programs. To examine their functional significance, we deleted 'hotspot' enhancer elements for Six2 and Wnt4. Removal of the distal enhancer for Six2 leads to a ~40% reduction in Six2 expression. When combined with a Six2 null allele, progeny display a premature depletion of nephron progenitors. Loss of the Wnt4 enhancer led to a significant reduction of Wnt4 expression in renal vesicles and a mildly hypoplastic kidney, a phenotype also enhanced in combination with a Wnt4 null mutation. To explore the regulatory landscape that supports proper target gene expression, we performed CTCF ChIP-seq to identify insulator-boundary regions. One such putative boundary lies between the Six2 and Six3 loci. Evidence for the functional significance of this boundary was obtained by deep sequencing of the radiation-induced Brachyrrhine (Br) mutant allele. We identified an inversion of the Six2/Six3 locus around the CTCF-bound boundary, removing Six2 from its distal enhancer regulation, but placed next to Six3 enhancer elements which support ectopic Six2 expression in the lens where Six3 is normally expressed. Six3 is now predicted to fall under control of the Six2 distal enhancer. Consistent with this view, we observed ectopic Six3 in nephron progenitors. 4C-seq supports the model for Six2 distal enhancer interactions in wild-type and Br/+ mouse kidneys. Together, these data expand our view of the regulatory genome and regulatory landscape underpinning mammalian nephrogenesis.


Subject(s)
Cell Differentiation/genetics , Gene Regulatory Networks , Nephrons/embryology , Organogenesis/genetics , Stem Cells/physiology , Transcription Factors/physiology , Animals , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Transcription Factors/genetics , Transcription Factors/isolation & purification , Wnt4 Protein/genetics , Wnt4 Protein/physiology
14.
Development ; 143(4): 595-608, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26884396

ABSTRACT

Nephron endowment is determined by the self-renewal and induction of a nephron progenitor pool established at the onset of kidney development. In the mouse, the related transcriptional regulators Six1 and Six2 play non-overlapping roles in nephron progenitors. Transient Six1 activity prefigures, and is essential for, active nephrogenesis. By contrast, Six2 maintains later progenitor self-renewal from the onset of nephrogenesis. We compared the regulatory actions of Six2 in mouse and human nephron progenitors by chromatin immunoprecipitation followed by DNA sequencing (ChIP-seq). Surprisingly, SIX1 was identified as a SIX2 target unique to the human nephron progenitors. Furthermore, RNA-seq and immunostaining revealed overlapping SIX1 and SIX2 activity in 16 week human fetal nephron progenitors. Comparative bioinformatic analysis of human SIX1 and SIX2 ChIP-seq showed each factor targeted a similar set of cis-regulatory modules binding an identical target recognition motif. In contrast to the mouse where Six2 binds its own enhancers but does not interact with DNA around Six1, both human SIX1 and SIX2 bind homologous SIX2 enhancers and putative enhancers positioned around SIX1. Transgenic analysis of a putative human SIX1 enhancer in the mouse revealed a transient, mouse-like, pre-nephrogenic, Six1 regulatory pattern. Together, these data demonstrate a divergence in SIX-factor regulation between mouse and human nephron progenitors. In the human, an auto/cross-regulatory loop drives continued SIX1 and SIX2 expression during active nephrogenesis. By contrast, the mouse establishes only an auto-regulatory Six2 loop. These data suggest differential SIX-factor regulation might have contributed to species differences in nephron progenitor programs such as the duration of nephrogenesis and the final nephron count.


Subject(s)
Gene Expression Regulation , Homeodomain Proteins/metabolism , Nephrons/cytology , Nerve Tissue Proteins/metabolism , Stem Cells/cytology , Transcription Factors/metabolism , Animals , Enhancer Elements, Genetic , Gene Regulatory Networks , Humans , Kidney/embryology , Kidney/metabolism , Mice, Transgenic , Models, Biological , Stem Cells/metabolism
15.
Semin Cell Dev Biol ; 36: 31-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25194660

ABSTRACT

The functional unit of the mammalian metanephric kidney is the nephron: a complex tubular structure dedicated to blood filtration and maintenance of several important physiological functions. Nephrons are assembled from a nephron-restricted pool of mesenchymal progenitors over an extensive developmental period that is completed prior to (human), or shortly after (mouse), birth. An appropriate balance in the expansion and commitment of nephron progenitors to nephron formation is essential for normal kidney function. Too few nephrons increase risk of kidney disease later in life while the failure of normal progenitor differentiation in Wilm's tumor patients leads to massive growth of a nephroblast population often necessitating surgical removal of the kidney. An inductive process within the metanephric mesenchyme leads to the formation of a pretubular aggregate which transitions into an epithelial renal vesicle: the precursor for nephron assembly. Growth, morphogenesis and patterning transform this simple cyst-like structure into a highly elongated mature nephron with distinct cell types positioned along a proximal (glomerular) to distal (connecting segment) axis of functional organization. This review discusses our current understanding of the specification, maintenance and commitment of nephron progenitors, and the regulatory processes that transform the renal vesicle into a nephron.


Subject(s)
Mesenchymal Stem Cells/cytology , Nephrons/cytology , Nephrons/embryology , Organogenesis/physiology , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Humans , Mice , Receptors, Notch/metabolism , Wilms Tumor/embryology , Wnt Proteins/metabolism , Wnt Signaling Pathway
16.
Nephrology (Carlton) ; 18(3): 177-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23279704

ABSTRACT

Nephrogenesis is dependent on the input of several transcriptional regulatory networks. However, the details of how these networks operate and converge to facilitate nephron progenitor specific programmes are largely unknown. To this end, recent studies have focused on identifying the precise regulatory mechanisms that modulate progenitor maintenance and induction. Continued focus on this area of research will help identify nephrogenic programmes which could be manipulated for therapeutic intervention of kidney disease.


Subject(s)
Cell Differentiation , Cell Proliferation , Nephrons/growth & development , Stem Cells/physiology , Animals , Gene Expression Regulation, Developmental , Humans , Nephrons/embryology , Nephrons/metabolism , Organogenesis , Signal Transduction , Stem Cells/metabolism , Transcription Factors/metabolism
17.
Dev Cell ; 23(3): 637-51, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22902740

ABSTRACT

A balance between Six2-dependent self-renewal and canonical Wnt signaling-directed commitment regulates mammalian nephrogenesis. Intersectional studies using chromatin immunoprecipitation and transcriptional profiling identified direct target genes shared by each pathway within nephron progenitors. Wnt4 and Fgf8 are essential for progenitor commitment; cis-regulatory modules flanking each gene are cobound by Six2 and ß-catenin and are dependent on conserved Lef/Tcf binding sites for activity. In vitro and in vivo analyses suggest that Six2 and Lef/Tcf factors form a regulatory complex that promotes progenitor maintenance while entry of ß-catenin into this complex promotes nephrogenesis. Alternative transcriptional responses associated with Six2 and ß-catenin cobinding events occur through non-Lef/Tcf DNA binding mechanisms, highlighting the regulatory complexity downstream of Wnt signaling in the developing mammalian kidney.


Subject(s)
Gene Regulatory Networks , Homeodomain Proteins/metabolism , Nephrons/cytology , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cells, Cultured , Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Nephrons/embryology , Nephrons/metabolism , Transcription Factors/genetics , beta Catenin/genetics
18.
Dev Biol ; 358(2): 318-30, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21871448

ABSTRACT

Podocytes help form the glomerular blood filtration barrier in the kidney and their injury or loss leads to renal disease. The Wilms' tumor suppressor-1 (Wt1) and the FoxC1/2 transcription factors, as well as Notch signaling, have been implicated as important regulators of podocyte fate. It is not known whether these factors work in parallel or sequentially on different gene targets, or as higher-order transcriptional complexes on common genes. Here, we use the zebrafish to demonstrate that embryos treated with morpholinos against wt1a, foxc1a, or the Notch transcriptional mediator rbpj develop fewer podocytes, as determined by wt1b, hey1 and nephrin expression, while embryos deficient in any two of these factors completely lack podocytes. From GST-pull-downs and co-immunoprecipitation experiments we show that Wt1a, Foxc1a, and Rbpj can physically interact with each other, whereas only Rbpj binds to the Notch intracellular domain (NICD). In transactivation assays, combinations of Wt1, FoxC1/2, and NICD synergistically induce the Hey1 promoter, and have additive or repressive effects on the Podocalyxin promoter, depending on dosage. Taken together, these data suggest that Wt1, FoxC1/2, and Notch signaling converge on common target genes where they physically interact to regulate a podocyte-specific gene program. These findings further our understanding of the transcriptional circuitry responsible for podocyte formation and differentiation during kidney development.


Subject(s)
Forkhead Transcription Factors/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Podocytes/cytology , Podocytes/metabolism , Receptors, Notch/metabolism , WT1 Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , DNA, Antisense/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Immunoglobulin J Recombination Signal Sequence-Binding Protein/antagonists & inhibitors , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Kidney/cytology , Kidney/embryology , Kidney/metabolism , Models, Biological , Promoter Regions, Genetic , Protein Interaction Domains and Motifs , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics , Zebrafish/genetics , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
19.
J Cell Biol ; 182(1): 77-88, 2008 Jul 14.
Article in English | MEDLINE | ID: mdl-18606852

ABSTRACT

Mitotic spindles are microtubule-based structures responsible for chromosome partitioning during cell division. Although the roles of microtubules and microtubule-based motors in mitotic spindles are well established, whether or not actin filaments (F-actin) and F-actin-based motors (myosins) are required components of mitotic spindles has long been controversial. Based on the demonstration that myosin-10 (Myo10) is important for assembly of meiotic spindles, we assessed the role of this unconventional myosin, as well as F-actin, in mitotic spindles. We find that Myo10 localizes to mitotic spindle poles and is essential for proper spindle anchoring, normal spindle length, spindle pole integrity, and progression through metaphase. Furthermore, we show that F-actin localizes to mitotic spindles in dynamic cables that surround the spindle and extend between the spindle and the cortex. Remarkably, although proper anchoring depends on both F-actin and Myo10, the requirement for Myo10 in spindle pole integrity is F-actin independent, whereas F-actin and Myo10 actually play antagonistic roles in maintenance of spindle length.


Subject(s)
Actin Cytoskeleton/metabolism , Myosins/metabolism , Spindle Apparatus/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/metabolism , Actins/metabolism , Animals , Cell Polarity , Cell Survival , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Female , Humans , Mitosis , Models, Biological , Myosins/chemistry , Myosins/deficiency , Protein Transport , Xenopus Proteins/chemistry , Xenopus Proteins/deficiency
20.
Dev Cell ; 11(2): 133-4, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16890151

ABSTRACT

Aurora-A kinases are highly conserved mitotic kinases required for cell division. The regulation of Aurora-A activity is less highly conserved and currently poorly understood. Work by Knoblich and coworkers in this issue of Developmental Cell identifies the conserved protein, Aurora Borealis (Bora), as a key regulator of Aurora-A activity during mitosis.


Subject(s)
Drosophila Proteins/metabolism , Mitosis/physiology , Protein Serine-Threonine Kinases/metabolism , Animals , Aurora Kinases , CDC2 Protein Kinase/metabolism , Drosophila
SELECTION OF CITATIONS
SEARCH DETAIL
...