Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Nat Commun ; 15(1): 3330, 2024 Apr 29.
Article En | MEDLINE | ID: mdl-38684656

Meiotic prophase progression is differently regulated in males and females. In males, pachytene transition during meiotic prophase is accompanied by robust alteration in gene expression. However, how gene expression is regulated differently to ensure meiotic prophase completion in males remains elusive. Herein, we identify HSF5 as a male germ cell-specific heat shock transcription factor (HSF) for meiotic prophase progression. Genetic analyzes and single-cell RNA-sequencing demonstrate that HSF5 is essential for progression beyond the pachytene stage under non-stress conditions rather than heat stress. Chromatin binding analysis in vivo and DNA-binding assays in vitro suggest that HSF5 binds to promoters in a subset of genes associated with chromatin organization. HSF5 recognizes a DNA motif different from typical heat shock elements recognized by other canonical HSFs. This study suggests that HSF5 is an atypical HSF that is required for the gene expression program for pachytene transition during meiotic prophase in males.


Heat Shock Transcription Factors , Meiotic Prophase I , Spermatogenesis , Mice , Male , Heat Shock Transcription Factors/metabolism , Mice, Knockout , Mice, Inbred C57BL , Testis/metabolism , Heat-Shock Response , Female
2.
Nat Commun ; 14(1): 6443, 2023 10 25.
Article En | MEDLINE | ID: mdl-37880249

Meiosis is differently regulated in males and females. In females, germ cells initiate meiosis within a limited time period in the fetal ovary and undergo a prolonged meiotic arrest until puberty. However, how meiosis initiation is coordinated with the cell cycle to coincide with S phase remains elusive. Here, we demonstrate that STRA8 binds to RB via the LXCXE motif. Mutation of the RB-binding site of STRA8 in female mice delays meiotic entry, which consequently delays progression of meiotic prophase and leads to precocious depletion of the oocyte pool. Single-cell RNA-sequencing analysis reveals that the STRA8-RB interaction is required for S phase entry and meiotic gene activation, ensuring precise timing of meiosis initiation in oocytes. Strikingly, the results suggest STRA8 could sequester RB from E2F during pre-meiotic G1/S transition. This study highlights the gene regulatory mechanisms underlying the female-specific mode of meiotic initiation in mice.


Adaptor Proteins, Signal Transducing , Meiosis , Animals , Female , Male , Mice , Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation , Germ Cells/metabolism , Sexual Maturation , Retinoblastoma Protein
3.
iScience ; 25(4): 104008, 2022 Apr 15.
Article En | MEDLINE | ID: mdl-35310947

Meiotic prophase I is a prolonged G2 phase that ensures the completion of numerous meiosis-specific chromosome events. During meiotic prophase I, homologous chromosomes undergo synapsis to facilitate meiotic recombination yielding crossovers. It remains largely elusive how homolog synapsis is temporally maintained and destabilized during meiotic prophase I. Here we show that FBXO47 is the stabilizer of the synaptonemal complex during male meiotic prophase I. Disruption of FBXO47 shows severe impact on homologous chromosome synapsis, meiotic recombination, and XY body formation, leading to male infertility. Notably, in the absence of FBXO47, although once homologous chromosomes are synapsed, the synaptonemal complex is precociously disassembled before progressing beyond pachytene. Remarkably, Fbxo47 KO spermatocytes remain in an earlier stage of meiotic prophase I and lack crossovers, despite apparently exhibiting diplotene-like chromosome morphology. We propose that FBXO47 plays a crucial role in preventing the synaptonemal complex from premature disassembly during cell cycle progression of meiotic prophase I.

4.
Curr Biol ; 32(7): 1485-1496.e4, 2022 04 11.
Article En | MEDLINE | ID: mdl-35196509

Developing animals absorb nutrients either through the placenta or from ingested food; however, the mechanisms by which embryos use external nutrients for individual organ morphogenesis remain to be elucidated. In this study, we assessed nutrient-dependent thyroid follicle morphogenesis in Xenopus laevis and investigated the role of secreted gastrointestinal (GI) hormones post-feeding. We found that feeding triggers thyroid follicle formation, and the thyroid cells showed transient inactivation of cell proliferation after feeding. In addition, the thyroid cells with multi-lumina were frequently observed in the fed tadpoles. The expression of the particular GI hormone incretin, glucose-dependent insulinotropic polypeptide (GIP), responded to feeding in the intestines of Xenopus tadpoles. Inhibition of dipeptidyl peptidase 4 (Dpp4), a degradative enzyme of incretin, increased the size of the thyroid follicles by facilitating follicular lumina connection, whereas inhibition of the sodium-glucose cotransporter (SGLT) reversed the effects of Dpp4 inhibition. Furthermore, injection of GIP peptide in unfed tadpoles initiated thyroid follicle formation-without requiring feeding-and injection of an incretin receptor antagonist suppressed follicle enlargement in the fed tadpoles. Lastly, GIP receptor knockout in neonatal mice showed smaller follicles in the thyroid, suggesting that the GI hormone-dependent thyroid morphogenesis is conserved in mammals. In conclusion, our study links external nutrients to thyroid morphogenesis and provides new insights into the function of GI hormone as a regulator of organ morphology in developing animals.


Gastrointestinal Hormones , Incretins , Animals , Dipeptidyl Peptidase 4/metabolism , Gastric Inhibitory Polypeptide/metabolism , Glucose/metabolism , Incretins/metabolism , Mammals , Mice , Morphogenesis , Thyroid Gland/metabolism
5.
Nat Commun ; 12(1): 3184, 2021 06 01.
Article En | MEDLINE | ID: mdl-34075040

During spermatogenesis, meiosis is accompanied by a robust alteration in gene expression and chromatin status. However, it remains elusive how the meiotic transcriptional program is established to ensure completion of meiotic prophase. Here, we identify a protein complex that consists of germ-cell-specific zinc-finger protein ZFP541 and its interactor KCTD19 as the key transcriptional regulators in mouse meiotic prophase progression. Our genetic study shows that ZFP541 and KCTD19 are co-expressed from pachytene onward and play an essential role in the completion of the meiotic prophase program in the testis. Furthermore, our ChIP-seq and transcriptome analyses identify that ZFP541 binds to and suppresses a broad range of genes whose function is associated with biological processes of transcriptional regulation and covalent chromatin modification. The present study demonstrates that a germ-cell specific complex that contains ZFP541 and KCTD19 promotes the progression of meiotic prophase towards completion in male mice, and triggers the reconstruction of the transcriptional network and chromatin organization leading to post-meiotic development.


Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Nuclear Proteins/metabolism , Pachytene Stage/genetics , Potassium Channels, Voltage-Gated/metabolism , Spermatids/cytology , Spermatogenesis/genetics , Transcription Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Chromatin Immunoprecipitation Sequencing , Chromosomal Proteins, Non-Histone/genetics , Disease Models, Animal , Female , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Infertility, Male/genetics , Male , Mice , Mice, Knockout , Nuclear Proteins/genetics , Oocytes/cytology , Oocytes/metabolism , Potassium Channels, Voltage-Gated/genetics , RNA-Seq , Spermatids/metabolism , Transcription Factors/genetics , Transcription, Genetic
6.
PLoS Genet ; 16(9): e1009048, 2020 09.
Article En | MEDLINE | ID: mdl-32931493

During meiotic prophase, sister chromatids are organized into axial element (AE), which underlies the structural framework for the meiotic events such as meiotic recombination and homolog synapsis. HORMA domain-containing proteins (HORMADs) localize along AE and play critical roles in the regulation of those meiotic events. Organization of AE is attributed to two groups of proteins: meiotic cohesins REC8 and RAD21L; and AE components SYCP2 and SYCP3. It has been elusive how these chromosome structural proteins contribute to the chromatin loading of HORMADs prior to AE formation. Here we newly generated Sycp2 null mice and showed that initial chromatin loading of HORMAD1 was mediated by meiotic cohesins prior to AE formation. HORMAD1 interacted not only with the AE components SYCP2 and SYCP3 but also with meiotic cohesins. Notably, HORMAD1 interacted with meiotic cohesins even in Sycp2-KO, and localized along cohesin axial cores independently of the AE components SYCP2 and SYCP3. Hormad1/Rad21L-double knockout (dKO) showed more severe defects in the formation of synaptonemal complex (SC) compared to Hormad1-KO or Rad21L-KO. Intriguingly, Hormad1/Rec8-dKO but not Hormad1/Rad21L-dKO showed precocious separation of sister chromatid axis. These findings suggest that meiotic cohesins REC8 and RAD21L mediate chromatin loading and the mode of action of HORMAD1 for synapsis during early meiotic prophase.


Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Animals , Chromatids/genetics , Chromatids/metabolism , Chromatin/metabolism , Chromosomes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Male , Meiosis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Phosphoproteins/genetics , Prophase/genetics , Spermatocytes/metabolism , Synaptonemal Complex/metabolism , Cohesins
7.
Sci Rep ; 10(1): 10094, 2020 06 22.
Article En | MEDLINE | ID: mdl-32572094

FZR1/CDH1 is an activator of Anaphase promoting complex/Cyclosome (APC/C), best known for its role as E3 ubiquitin ligase that drives the cell cycle. APC/C activity is regulated by CDK-mediated phosphorylation of FZR1 during mitotic cell cycle. Although the critical role of FZR1 phosphorylation has been shown mainly in yeast and in vitro cell culture studies, its biological significance in mammalian tissues in vivo remained elusive. Here, we examined the in vivo role of FZR1 phosphorylation using a mouse model, in which non-phosphorylatable substitutions were introduced in the putative CDK-phosphorylation sites of FZR1. Although ablation of FZR1 phosphorylation did not show substantial consequences in mouse somatic tissues, it led to severe testicular defects resulting in male infertility. In the absence of FZR1 phosphorylation, male juvenile germ cells entered meiosis normally but failed to enter meiosis II or form differentiated spermatids. In aged testis, male mutant germ cells were overall abolished, showing Sertoli cell-only phenotype. In contrast, female mutants showed apparently normal progression of meiosis. The present study demonstrated that phosphorylation of FZR1 is required for temporal regulation of APC/C activity at meiosis II entry, and for maintenance of spermatogonia, which raised an insight into the sexual dimorphism of FZR1-regulation in germ cells.


Cdh1 Proteins/metabolism , Meiosis/physiology , Anaphase-Promoting Complex-Cyclosome/metabolism , Animals , Cdh1 Proteins/physiology , Cell Cycle Proteins/metabolism , Gene Knock-In Techniques/methods , Germ Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Spermatogenesis/physiology , Spermatogonia/metabolism , Ubiquitin-Protein Ligases/metabolism
8.
Cell Rep ; 31(8): 107686, 2020 05 26.
Article En | MEDLINE | ID: mdl-32460033

Meiotic recombination is critical for genetic exchange and generation of chiasmata that ensures faithful chromosome segregation during meiosis I. Meiotic recombination is initiated by DNA double-strand break (DSB) followed by multiple processes of DNA repair. The exact mechanisms for how recombinases localize to DSB remain elusive. Here, we show that C19orf57/4930432K21Rik/BRME1 is a player for meiotic recombination in mice. C19orf57/4930432K21Rik/BRME1 associates with single-stranded DNA (ssDNA) binding proteins, BRCA2 and MEILB2/HSF2BP, which are critical recruiters of recombinases onto DSB sites. Disruption of C19orf57/4930432K21Rik/BRME1 shows severe impact on DSB repair and male fertility. Remarkably, removal of ssDNA binding proteins from DSB sites is delayed, and reciprocally, the loading of RAD51 and DMC1 onto resected ssDNA is impaired in Brme1 knockout (KO) spermatocytes. We propose that C19orf57/4930432K21Rik/BRME1 modulates localization of recombinases to meiotic DSB sites through the interaction with the BRCA2-MEILB2/HSF2BP complex during meiotic recombination.


Cell Cycle Proteins/metabolism , Homologous Recombination/genetics , Meiosis/genetics , Phosphate-Binding Proteins/metabolism , Rad51 Recombinase/metabolism , Spermatocytes/metabolism , Spermatogenesis/genetics , Animals , DNA Breaks, Double-Stranded , Male , Mice
9.
Dev Cell ; 52(4): 429-445.e10, 2020 02 24.
Article En | MEDLINE | ID: mdl-32032549

The mechanisms regulating meiotic initiation in mammals are enigmatic. It is known that retinoic acid (RA) signaling plays a pivotal role during meiotic initiation. STRA8, which is expressed in response to RA, is thought to be a key factor promoting meiotic initiation. However, the specific role of STRA8 in meiotic initiation has remained elusive. Here, we identified MEIOSIN as a germ-cell-specific factor that associates with STRA8. MEIOSIN, like STRA8, is expressed in response to RA and plays an essential role in meiotic initiation in both males and females. Functional analyses revealed that MEIOSIN acts as a transcription factor together with STRA8, and that both factors are critical for driving meiotic gene activation. Furthermore, temporally restricted expression of MEIOSIN leads to meiotic entry decision during spermatogenesis. The present study demonstrates that MEIOSIN, in collaboration with STRA8, plays a central role in regulating the mitosis to meiosis germ cell fate decision in mammals.


Adaptor Proteins, Signal Transducing/physiology , Cell Cycle , Gene Expression Regulation , Germ Cells/physiology , Meiosis , Mitosis , Transcription Factors/physiology , Animals , Cell Differentiation , Female , Germ Cells/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Spermatogenesis
10.
Dev Biol ; 427(1): 121-130, 2017 07 01.
Article En | MEDLINE | ID: mdl-28478097

In kidney development, connection of the nephric duct (ND) to the cloaca and subsequent sprouting of the ureteric bud (UB) from the ND are important for urinary exit tract formation. Although the roles of Ret signaling are well established, it remains unclear how intracellular cytoskeletal proteins regulate these morphogenetic processes. Myh9 and Myh10 encode two different non-muscle myosin II heavy chains, and Myh9 mutations in humans are implicated in congenital kidney diseases. Here we report that ND/UB lineage-specific deletion of Myh9/Myh10 in mice caused severe hydroureter/hydronephrosis at birth. At mid-gestation, the mutant ND/UB epithelia exhibited aberrant basal protrusion and ectopic UB formation, which likely led to misconnection of the ureter to the bladder. In addition, the mutant epithelia exhibited apical extrusion followed by massive apoptosis in the lumen, which could be explained by reduced apical constriction and intercellular adhesion mediated by E-cadherin. These phenotypes were not ameliorated by genetic reduction of the tyrosine kinase receptor Ret. In contrast, ERK was activated in the mutant cells and its chemical inhibition partially ameliorated the phenotypes. Thus, myosin II is essential for maintaining the apicobasal integrity of the developing kidney epithelia independently of Ret signaling.


Epithelium/abnormalities , Kidney/embryology , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIB/metabolism , Ureter/abnormalities , Urinary Bladder/abnormalities , Animals , Animals, Newborn , Dogs , Epithelium/metabolism , Gene Expression Regulation, Developmental , Immunohistochemistry , In Situ Hybridization , Kidney/metabolism , Madin Darby Canine Kidney Cells , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Nephrons/abnormalities , Nephrons/metabolism , Nonmuscle Myosin Type IIA/genetics , Nonmuscle Myosin Type IIB/genetics , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Ureter/metabolism , Urinary Bladder/metabolism
11.
Sci Rep ; 5: 15676, 2015 Oct 29.
Article En | MEDLINE | ID: mdl-26511275

The mammalian kidney develops from reciprocal interactions between the metanephric mesenchyme and ureteric bud, the former of which contains nephron progenitors. The third lineage, the stroma, fills up the interstitial space and is derived from distinct progenitors that express the transcription factor Foxd1. We showed previously that deletion of the nuclear factor Sall1 in nephron progenitors leads to their depletion in mice. However, Sall1 is expressed not only in nephron progenitors but also in stromal progenitors. Here we report that specific Sall1 deletion in stromal progenitors leads to aberrant expansion of nephron progenitors, which is in sharp contrast with a nephron progenitor-specific deletion. The mutant mice also exhibited cystic kidneys after birth and died before adulthood. We found that Decorin, which inhibits Bmp-mediated nephron differentiation, was upregulated in the mutant stroma. In contrast, the expression of Fat4, which restricts nephron progenitor expansion, was reduced mildly. Furthermore, the Sall1 protein binds to many stroma-related gene loci, including Decorin and Fat4. Thus, the expression of Sall1 in stromal progenitors restricts the excessive expansion of nephron progenitors in a non-cell autonomous manner, and Sall1-mediated regulation of Decorin and Fat4 might at least partially underlie the pathogenesis.


Cell Differentiation , Gene Expression Regulation , Nephrons/metabolism , Stem Cells/metabolism , Transcription Factors/biosynthesis , Animals , Cadherins/biosynthesis , Cadherins/genetics , Decorin/biosynthesis , Decorin/genetics , Mice , Mice, Mutant Strains , Nephrons/pathology , Stem Cells/pathology , Transcription Factors/genetics
12.
J Am Soc Nephrol ; 26(5): 1081-91, 2015 May.
Article En | MEDLINE | ID: mdl-25168025

The kidney develops from reciprocal interactions between the metanephric mesenchyme and ureteric bud. The mesenchyme transforms into epithelia and forms complicated nephron structures, whereas the ureteric bud extends its pre-existing epithelial ducts. Although the roles are well established for extracellular stimuli, such as Wnt and Notch, it is unclear how the intracellular cytoskeleton regulates these morphogenetic processes. Myh9 and Myh10 encode nonmuscle myosin II heavy chains, and Myh9 mutations in humans are implicated in congenital kidney diseases and focal segmental glomerulosclerosis in adults. Here, we analyzed the roles of Myh9 and Myh10 in the developing kidney. Ureteric bud-specific depletion of Myh9 resulted in no apparent phenotypes, whereas mesenchyme-specific Myh9 deletion caused proximal tubule dilations and renal failure. Mesenchyme-specific Myh9/Myh10 mutant mice died shortly after birth and showed a severe defect in nephron formation. The nascent mutant nephrons failed to form a continuous lumen, which likely resulted from impaired apical constriction of the elongating tubules. In addition, nephron progenitors lacking Myh9/Myh10 or the possible interactor Kif26b were less condensed at midgestation and reduced at birth. Taken together, nonmuscle myosin II regulates the morphogenesis of immature nephrons derived from the metanephric mesenchyme and the maintenance of nephron progenitors. Our data also suggest that Myh9 deletion in mice results in failure to maintain renal tubules but not in glomerulosclerosis.


Morphogenesis , Myosin Heavy Chains/physiology , Nephrons/embryology , Nonmuscle Myosin Type IIA/physiology , Nonmuscle Myosin Type IIB/physiology , Animals , Animals, Newborn , Mesoderm/physiology , Mice, Inbred C57BL , Nephrons/metabolism , Protein Isoforms/metabolism
13.
J Am Soc Nephrol ; 24(8): 1242-9, 2013 Jul.
Article En | MEDLINE | ID: mdl-23641053

Islet1 (Isl1) is a transcription factor transiently expressed in a subset of heart and limb progenitors. During studies of limb development, conditional Isl1 deletion produced unexpected kidney abnormalities. Here, we studied the renal expression of Isl1 and whether it has a role in kidney development. In situ hybridization revealed Isl1 expression in the mesenchymal cells surrounding the base of the ureteric bud in mice. Conditional deletion of Isl1 caused kidney agenesis or hypoplasia and hydroureter, a phenotype resembling human congenital anomalies of the kidney and urinary tract (CAKUT). The absence of Isl1 led to ectopic branching of the ureteric bud out from the nephric duct or to the formation of accessory buds, both of which could lead to obstruction of the ureter-bladder junction and consequent hydroureter. The abnormal elongation and poor branching of the ureteric buds were the likely causes of the kidney agenesis or hypoplasia. Furthermore, the lack of Isl1 reduced the expression of Bmp4, a gene implicated in the CAKUT-like phenotype, in the metanephric region before ureteric budding. In conclusion, Isl1 is essential for proper development of the kidney and ureter by repressing the aberrant formation of the ureteric bud. These observations call for further studies to investigate whether Isl1 may be a causative gene for human CAKUT.


Congenital Abnormalities/genetics , Gene Expression Regulation, Developmental/genetics , Kidney Diseases/congenital , LIM-Homeodomain Proteins/genetics , Organogenesis/genetics , Transcription Factors/genetics , Ureter/abnormalities , Animals , Gene Deletion , Kidney/abnormalities , Kidney Diseases/genetics , Mice , Ureter/metabolism
14.
Nat Commun ; 4: 1398, 2013.
Article En | MEDLINE | ID: mdl-23360989

Most kidney nephron components, including glomeruli and renal tubules, derive from the metanephric mesenchyme. The overall differentiation into each component finishes at birth, but the molecular events linking the perinatal and adult kidneys remain elusive. Dullard was cloned from Xenopus kidneys, and encodes a phosphatase that negatively regulates BMP signalling. Here we report that Dullard deletion in the murine metanephric mesenchyme leads to failure of nephron maintenance after birth, resulting in lethality before adulthood. The nephron components are lost by massive apoptosis within 3 weeks after birth, leading to formation of a large hollow with a thin-layered cortex and medulla. Phosphorylated Smad1/5/8 is upregulated in the mutant nephrons, probably through cell-autonomous inhibitory effects of Dullard on BMP signalling. Importantly, administration of the BMP receptor kinase inhibitor LDN-193189 partially rescued the defects caused by Dullard deletion. Thus, Dullard keeps BMP signalling at an appropriate level, which is required for nephron maintenance in the postnatal period.


Bone Morphogenetic Proteins/metabolism , Nephrons/enzymology , Phosphoprotein Phosphatases/metabolism , Signal Transduction , Aging/metabolism , Animals , Animals, Newborn , Apoptosis , Blotting, Western , Cell Line , Mice , Mice, Mutant Strains , Nephrons/embryology , Nephrons/pathology , Phosphoprotein Phosphatases/deficiency , Staining and Labeling
15.
Pediatr Nephrol ; 26(9): 1463-7, 2011 Sep.
Article En | MEDLINE | ID: mdl-21336811

The kidney is formed by a reciprocally inductive interaction between two precursor tissues, the metanephric mesenchyme and the ureteric bud. This interaction can be divided into three processes: attraction of the ureteric bud toward the mesenchyme, maintenance of the mesenchyme in an undifferentiated state versus transition to an epithelial state, and further differentiation into multiple epithelial lineages, such as glomeruli and renal tubules. In this review we describe our recent findings related to each process. A mesenchymal nuclear zinc finger protein, Sall1, controls ureteric bud attraction by regulating a novel kinesin, Kif26b. The Sall1 gene is highly expressed in multipotent nephron progenitors in the mesenchyme, and these cells can partially reconstitute a three-dimensional structure in organ cultures following Wnt4 stimulation. While Notch2 is required for further differentiation of proximal nephron structures, ectopic Notch2 activation in the embryonic kidney depletes nephron progenitors, suggesting that Notch2 stabilizes--rather than dictates--nephron fate by shutting down the maintenance of undifferentiated progenitor cells.


Mesenchymal Stem Cells/physiology , Nephrons/embryology , Animals , Cell Differentiation , Cell Lineage , Gene Expression Regulation, Developmental , Humans , Mesenchymal Stem Cells/metabolism , Nephrons/metabolism , Organogenesis , Signal Transduction
16.
Proc Natl Acad Sci U S A ; 107(20): 9240-5, 2010 May 18.
Article En | MEDLINE | ID: mdl-20439720

The kidney develops through reciprocal interactions between two precursor tissues: the metanephric mesenchyme and the ureteric bud. We previously demonstrated that the zinc finger protein Sall1 is essential for ureteric bud attraction toward the mesenchyme. Here, we show that Kif26b, a kinesin family gene, is a downstream target of Sall1 and that disruption of this gene causes kidney agenesis because of impaired ureteric bud attraction. In the Kif26b-null metanephros, compact adhesion between mesenchymal cells adjacent to the ureteric buds and the polarized distribution of integrin alpha8 were impaired, resulting in failed maintenance of Gdnf, a critical ureteric bud attractant. Overexpression of Kif26b in vitro caused increased cell adhesion through interactions with nonmuscle myosin. Thus, Kif26b is essential for kidney development because it regulates the adhesion of mesenchymal cells in contact with ureteric buds.


Cell Adhesion/physiology , Kidney/embryology , Kinesins/metabolism , Mesoderm/physiology , Animals , Blotting, Southern , Cloning, Molecular , DNA Primers/genetics , Female , Immunohistochemistry , In Situ Hybridization , Kidney/cytology , Kinesins/genetics , Mass Spectrometry , Mice , Mice, Inbred C57BL , Transfection
17.
J Am Soc Nephrol ; 21(5): 803-10, 2010 May.
Article En | MEDLINE | ID: mdl-20299358

Successive activation of Wnt4 and Notch2 generates nephrons from the metanephric mesenchyme. Mesenchymal-to-epithelial transition requires Wnt4, and normal development of the proximal nephron (epithelia of glomeruli and proximal tubules) requires Notch2. It is unknown, however, whether Notch2 dictates the fate of the proximal nephron directly. Here, we generated a mutant strain of mice with activated Notch2 in Six2-containing nephron progenitor cells of the metanephric mesenchyme. Notch2 activation did not skew the cell fate toward the proximal nephron but resulted in severe kidney dysgenesis and depletion of Six2-positive progenitors. We observed ectopic expression of Wnt4 and premature tubule formation, similar to the phenotype of Six2-deficient mice. Activation of Notch2 in the progenitor cells suppressed Pax2, an upstream regulator of Six2, possibly through Hesr genes. Taken together, these data suggest that a positive feedback loop exists between Notch2 and Wnt4, and that Notch2 stabilizes, rather than dictates, nephron fate by shutting down the maintenance of undifferentiated progenitor cells, thereby depleting this population.


Homeodomain Proteins/metabolism , Nephrons/embryology , Receptor, Notch2/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle Proteins/metabolism , Integrases/metabolism , Mice , Nephrons/metabolism , PAX2 Transcription Factor/metabolism , Wnt Proteins/metabolism , Wnt4 Protein
18.
Genesis ; 48(3): 207-12, 2010 Mar.
Article En | MEDLINE | ID: mdl-20127799

Sall1 is expressed in the metanephric mesenchyme in the developing kidney, and mice deficient in Sall1 show kidney agenesis or dysgenesis. Sall1 is also expressed elsewhere, including in the limb buds, anus, heart, and central nervous system. Dominant-negative mutations of Sall1 in mice and humans lead to developmental defects in these organs. Here, we generated a mouse line expressing tamoxifen-inducible Cre recombinase (CreER(T2)) under the control of the endogenous Sall1 promoter. Upon tamoxifen treatment, these mice showed genomic recombination in the tissues where endogenous Sall1 is expressed. When CreER(T2) mice were crossed with the floxed Sall1 allele, tamoxifen administration during gestation led to a significant decrease in Sall1 expression and small kidneys at birth, suggesting that Sall1 functions were disrupted. Furthermore, Sall1 expression in the kidney was significantly reduced by neonatal tamoxifen treatment. The Sall1CreER(T2) mouse is a valuable tool for in vivo time-dependent and region-specific knockout and overexpression studies.


Integrases/genetics , Kidney/metabolism , Mesoderm/metabolism , Transcription Factors/genetics , Animals , Animals, Newborn , Embryo, Mammalian/drug effects , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Gene Targeting/methods , Humans , Immunohistochemistry , In Situ Hybridization , Integrases/metabolism , Kidney/embryology , Kidney/growth & development , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mesoderm/embryology , Mesoderm/growth & development , Mice , Mice, Knockout , Mice, Transgenic , Models, Genetic , Promoter Regions, Genetic/genetics , Tamoxifen/pharmacology
19.
J Biochem ; 147(3): 445-50, 2010 Mar.
Article En | MEDLINE | ID: mdl-20053786

Human SALL1 is a homologue of the Drosophila region-specific homeotic gene sal, and is also known as a causative gene for Townes-Brocks syndrome, which is characterized by multi-organ malformations. We previously demonstrated that mouse Sall1 plays a crucial role in ureteric bud invasion during kidney development, and possibly in nephron progenitor cells in the metanephric mesenchyme. To gain insights into the Sall1 functions in the kidney and other tissues, we generated R26Sall1 mice, in which Rosa26 locus stop sequences flanked by two loxP sites were located upstream of the Sall1 cDNA. This allele allowed exogenous Sall1 expression in a Cre recombinase-dependent manner. R26Sall1 mice were first crossed with CAGCre mice, which expressed Cre recombinase ubiquitously during embryogenesis. Mice expressing Sall1 ubiquitously were smaller in size compared with mice of other genotypes. We then crossed R26Sall1 mice with Six2Cre mice expressing Cre recombinase in the metanephric mesenchyme during kidney development. However, no kidney defects were observed. Taken together, overexpression of Sall1 does not affect kidney development, but does lead to a reduced body weight, suggesting that the optimal dosage of Sall1 is required for normal mouse development.


Body Weight , Kidney/embryology , Transcription Factors , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Animals , Anus, Imperforate/genetics , Anus, Imperforate/metabolism , Ear/abnormalities , Gene Expression Regulation, Developmental , Humans , Kidney/cytology , Kidney/metabolism , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/metabolism , Mice , Mice, Transgenic , Transcription Factors/biosynthesis , Transcription Factors/genetics
20.
Stem Cells ; 27(4): 796-805, 2009 Apr.
Article En | MEDLINE | ID: mdl-19350679

Sall4 is a mouse homolog of a causative gene of the autosomal dominant disorder Okihiro syndrome. We previously showed that the absence of Sall4 leads to lethality during peri-implantation and that Sall4-null embryonic stem (ES) cells proliferate poorly with intact pluripotency when cultured on feeder cells. Here, we report that, in the absence of feeder cells, Sall4-null ES cells express the trophectoderm marker Cdx2, but are maintained for a long period in an undifferentiated state with minimally affected Oct3/4 expression. Feeder-free Sall4-null ES cells contribute solely to the inner cell mass and epiblast in vivo, indicating that these cells still retain pluripotency and do not fully commit to the trophectoderm. These phenotypes could arise from derepression of the Cdx2 promoter, which is normally suppressed by Sall4 and the Mi2/NuRD HDAC complex. However, proliferation was impaired and G1 phase prolonged in the absence of Sall4, suggesting another role for Sall4 in cell cycle control. Although Sall1, also a Sall family gene, is known to genetically interact with Sall4 in vivo, Sall1-null ES cells have no apparent defects and no exacerbation is observed in ES cells lacking both Sall1 and Sall4, compared with Sall4-null cells. This suggests a unique role for Sall4 in ES cells. Thus, though Sall4 does not contribute to the central machinery of the pluripotency, it stabilizes ES cells by repressing aberrant trophectoderm gene expression.


Cell Differentiation/physiology , DNA-Binding Proteins/physiology , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental/physiology , Pluripotent Stem Cells/physiology , Transcription Factors/physiology , Animals , CDX2 Transcription Factor , Cell Cycle/genetics , Embryonic Development/genetics , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunohistochemistry , Immunoprecipitation , Mice , Microscopy, Confocal , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction
...