Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Front Cell Dev Biol ; 11: 1233317, 2023.
Article En | MEDLINE | ID: mdl-37727504

Kidney formation and nephrogenesis are controlled by precise spatiotemporal gene expression programs, which are coordinately regulated by cell-cycle, cell type-specific transcription factors and epigenetic/chromatin regulators. However, the roles of epigenetic/chromatin regulators in kidney development and disease remain poorly understood. In this study, we investigated the impact of deleting the chromatin remodeling factor Smarca4 (Brg1), a human Wilms tumor-associated gene, in Wnt4-expressing cells. Smarca4 deficiency led to severe tubular defects and a shortened medulla. Through unbiased single-cell RNA sequencing analyses, we identified multiple types of Wnt4 Cre-labeled interstitial cells, along with nephron-related cells. Smarca4 deficiency increased interstitial cells but markedly reduced tubular cells, resulting in cells with mixed identity and elevated expression of cell-cycle regulators and genes associated with extracellular matrix and epithelial-to-mesenchymal transition/fibrosis. We found that Smarca4 loss induced a significant upregulation of the oncogene Pttg1 and hyperproliferation of Wnt4 Cre-labeled cells. These changes in the cellular state could hinder the cellular transition into characteristic tubular structures, eventually leading to fibrosis. In conclusion, our findings shed light on novel cell types and genes associated with Wnt4 Cre-labeled cells and highlight the critical role of Smarca4 in regulating tubular cell differentiation and the expression of the cancer-causing gene Pttg1 in the kidney. These findings may provide valuable insights into potential therapeutic strategies for renal cell carcinoma resulting from SMARCA4 deficiency.

2.
Nucleic Acids Res ; 50(18): 10343-10359, 2022 10 14.
Article En | MEDLINE | ID: mdl-36130284

Eya1 is critical for establishing and maintaining nephron progenitor cells (NPCs). It belongs to a family of proteins called phosphatase-transcriptional activators but without intrinsic DNA-binding activity. However, the spectrum of the Eya1-centered networks is underexplored. Here, we combined transcriptomic, genomic and proteomic approaches to characterize gene regulation by Eya1 in the NPCs. We identified Eya1 target genes, associated cis-regulatory elements and partner proteins. Eya1 preferentially occupies promoter sequences and interacts with general transcription factors (TFs), RNA polymerases, different types of TFs, chromatin-remodeling factors with ATPase or helicase activity, and DNA replication/repair proteins. Intriguingly, we identified REST-binding motifs in 76% of Eya1-occupied sites without H3K27ac-deposition, which were present in many Eya1 target genes upregulated in Eya1-deficient NPCs. Eya1 copurified REST-interacting chromatin-remodeling factors, histone deacetylase/lysine demethylase, and corepressors. Coimmunoprecipitation validated physical interaction between Eya1 and Rest/Hdac1/Cdyl/Hltf in the kidneys. Collectively, our results suggest that through interactions with chromatin-remodeling factors and specialized DNA-binding proteins, Eya1 may modify chromatin structure to facilitate the assembly of regulatory complexes that regulate transcription positively or negatively. These findings provide a mechanistic basis for how Eya1 exerts its activity by forming unique multiprotein complexes in various biological processes to maintain the cellular state of NPCs.


Intracellular Signaling Peptides and Proteins/metabolism , Nephrons/cytology , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Adenosine Triphosphatases/genetics , Animals , Chromatin/genetics , Co-Repressor Proteins , DNA-Binding Proteins/genetics , Histone Deacetylases/metabolism , Mice , Multiprotein Complexes/genetics , Nephrons/metabolism , Phosphoric Monoester Hydrolases/genetics , Proteomics , Transcription Factors, General/genetics
3.
Front Cell Dev Biol ; 10: 815249, 2022.
Article En | MEDLINE | ID: mdl-35178390

The vertebrate Six1 and Six2 arose by gene duplication from the Drosophila sine oculis and have since diverged in their developmental expression patterns. Both genes are expressed in nephron progenitors of human fetal kidneys, and mutations in SIX1 or SIX2 cause branchio-oto-renal syndrome or renal hypodysplasia respectively. Since ∼80% of SIX1 target sites are shared by SIX2, it is speculated that SIX1 and SIX2 may be functionally interchangeable by targeting common downstream genes. In contrast, in mouse kidneys, Six1 expression in the metanephric mesenchyme lineage overlaps with Six2 only transiently, while Six2 expression is maintained in the nephron progenitors throughout development. This non-overlapping expression between Six1 and Six2 in mouse nephron progenitors promoted us to examine if Six1 can replace Six2. Surprisingly, forced expression of Six1 failed to rescue Six2-deficient kidney phenotype. We found that Six1 mediated Eya1 nuclear translocation and inhibited premature epithelialization of the progenitors but failed to rescue the proliferation defects and cell death caused by Six2-knockout. Genome-wide binding analyses showed that Six1 selectively occupied a small subset of Six2 target sites, but many Six2-bound loci crucial to the renewal and differentiation of nephron progenitors lacked Six1 occupancy. Altogether, these data indicate that Six1 cannot substitute Six2 to drive nephrogenesis in mouse kidneys, thus demonstrating that the difference in physiological roles of Six1 and Six2 in kidney development stems from both transcriptional regulations of the genes and divergent biochemical properties of the proteins.

4.
J Am Soc Nephrol ; 32(11): 2815-2833, 2021 11.
Article En | MEDLINE | ID: mdl-34716243

BACKGROUND: Eya1 is a critical regulator of nephron progenitor cell specification and interacts with Six2 to promote NPC self-renewal. Haploinsufficiency of these genes causes kidney hypoplasia. However, how the Eya1-centered network operates remains unknown. METHODS: We engineered a 2×HA-3×Flag-Eya1 knock-in mouse line and performed coimmunoprecipitation with anti-HA or -Flag to precipitate the multitagged-Eya1 and its associated proteins. Loss-of-function, transcriptome profiling, and genome-wide binding analyses for Eya1's interacting chromatin-remodeling ATPase Brg1 were carried out. We assayed the activity of the cis-regulatory elements co-occupied by Brg1/Six2 in vivo. RESULTS: Eya1 and Six2 interact with the Brg1-based SWI/SNF complex during kidney development. Knockout of Brg1 results in failure of metanephric mesenchyme formation and depletion of nephron progenitors, which has been linked to loss of Eya1 expression. Transcriptional profiling shows conspicuous downregulation of important regulators for nephrogenesis in Brg1-deficient cells, including Lin28, Pbx1, and Dchs1-Fat4 signaling, but upregulation of podocyte lineage, oncogenic, and cell death-inducing genes, many of which Brg1 targets. Genome-wide binding analysis identifies Brg1 occupancy to a distal enhancer of Eya1 that drives nephron progenitor-specific expression. We demonstrate that Brg1 enrichment to two distal intronic enhancers of Pbx1 and a proximal promoter region of Mycn requires Six2 activity and that these Brg1/Six2-bound enhancers govern nephron progenitor-specific expression in response to Six2 activity. CONCLUSIONS: Our results reveal an essential role for Brg1, its downstream pathways, and its interaction with Eya1-Six2 in mediating the fine balance among the self-renewal, differentiation, and survival of nephron progenitors.


Chromatin Assembly and Disassembly , DNA Helicases/physiology , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nephrons/cytology , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Protein Tyrosine Phosphatases/metabolism , Stem Cells/cytology , Transcription Factors/metabolism , Transcription Factors/physiology , Animals , Base Sequence , Cell Differentiation , Cell Self Renewal , Chromatin Immunoprecipitation , Gene Knock-In Techniques , HEK293 Cells , Homeodomain Proteins/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Kidney/embryology , Mesoderm/cytology , Mesoderm/metabolism , Mice , Multiprotein Complexes , Nuclear Proteins/genetics , Protein Interaction Mapping , Protein Tyrosine Phosphatases/genetics , Stem Cells/metabolism , Transcription Factors/genetics , Transcriptome
5.
Dev Dyn ; 250(10): 1450-1462, 2021 10.
Article En | MEDLINE | ID: mdl-33715274

BACKGROUND: Eya2 expression during mouse development has been studied by in situ hybridization and it has been shown to be involved skeletal muscle development and limb formation. Here, we generated Eya2 knockout (Eya2- ) and a lacZ knockin reporter (Eya2lacZ ) mice and performed a detailed expression analysis for Eya2lacZ at different developmental stages to trace Eya2lacZ -positive cells in Eya2-null mice. We describe that Eya2 is not only expressed in cranial sensory and dorsal root ganglia, retina and olfactory epithelium, and somites as previously reported, but also Eya2 is specifically detected in other organs during mouse development. RESULTS: We found that Eya2 is expressed in ocular and trochlear motor neurons. In the inner ear, Eya2lacZ is specifically expressed in differentiating hair cells in both vestibular and cochlear sensory epithelia of the inner ear and Eya2-/- or Eya2lacZ/lacZ mice displayed mild hearing loss. Furthermore, we detected Eya2 expression during both salivary gland and thymus development and Eya2-null mice had a smaller thymus. CONCLUSIONS: As Eya2 is coexpressed with other members of the Eya family genes, these results together highlight that Eya2 as a potential regulator may act synergistically with other Eya genes to regulate the differentiation of the inner ear sensory hair cells and the formation of the salivary gland and thymus.


Ear, Inner/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Hearing Loss/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Animals , Cell Differentiation/physiology , Ear, Inner/embryology , Hearing Loss/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Proteins/genetics , Protein Tyrosine Phosphatases/genetics
6.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Article En | MEDLINE | ID: mdl-33723076

Specification of Sox2+ proneurosensory progenitors within otic ectoderm is a prerequisite for the production of sensory cells and neurons for hearing. However, the underlying molecular mechanisms driving this lineage specification remain unknown. Here, we show that the Brg1-based SWI/SNF chromatin-remodeling complex interacts with the neurosensory-specific transcriptional regulators Eya1/Six1 to induce Sox2 expression and promote proneurosensory-lineage specification. Ablation of the ATPase-subunit Brg1 or both Eya1/Six1 results in loss of Sox2 expression and lack of neurosensory identity, leading to abnormal apoptosis within the otic ectoderm. Brg1 binds to two of three distal 3' Sox2 enhancers occupied by Six1, and Brg1-binding to these regions depends on Eya1-Six1 activity. We demonstrate that the activity of these Sox2 enhancers in otic neurosensory cells specifically depends on binding to Six1. Furthermore, genome-wide and transcriptome profiling indicate that Brg1 may suppress apoptotic factor Map3k5 to inhibit apoptosis. Together, our findings reveal an essential role for Brg1, its downstream pathways, and their interactions with Six1/Eya1 in promoting proneurosensory fate induction in the otic ectoderm and subsequent neuronal lineage commitment and survival of otic cells.


Chromatin Assembly and Disassembly , Chromatin/genetics , Chromatin/metabolism , Ectoderm/embryology , Ectoderm/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Neurons/cytology , Neurons/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
7.
Nucleic Acids Res ; 48(6): 2880-2896, 2020 04 06.
Article En | MEDLINE | ID: mdl-31956913

The transcription factor Six1 is essential for induction of sensory cell fate and formation of auditory sensory epithelium, but how it activates gene expression programs to generate distinct cell-types remains unknown. Here, we perform genome-wide characterization of Six1 binding at different stages of auditory sensory epithelium development and find that Six1-binding to cis-regulatory elements changes dramatically at cell-state transitions. Intriguingly, Six1 pre-occupies enhancers of cell-type-specific regulators and effectors before their expression. We demonstrate in-vivo cell-type-specific activity of Six1-bound novel enhancers of Pbx1, Fgf8, Dusp6, Vangl2, the hair-cell master regulator Atoh1 and a cascade of Atoh1's downstream factors, including Pou4f3 and Gfi1. A subset of Six1-bound sites carry consensus-sequences for its downstream factors, including Atoh1, Gfi1, Pou4f3, Gata3 and Pbx1, all of which physically interact with Six1. Motif analysis identifies RFX/X-box as one of the most significantly enriched motifs in Six1-bound sites, and we demonstrate that Six1-RFX proteins cooperatively regulate gene expression through binding to SIX:RFX-motifs. Six1 targets a wide range of hair-bundle regulators and late Six1 deletion disrupts hair-bundle polarity. This study provides a mechanistic understanding of how Six1 cooperates with distinct cofactors in feedforward loops to control lineage-specific gene expression programs during progressive differentiation of the auditory sensory epithelium.


Cell Differentiation/genetics , Cell Lineage/genetics , Epithelium/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/cytology , Hair Cells, Auditory/metabolism , Homeodomain Proteins/metabolism , Base Sequence , Cell Polarity , Consensus Sequence , DNA/metabolism , Dual Specificity Phosphatase 6/genetics , Dual Specificity Phosphatase 6/metabolism , Enhancer Elements, Genetic/genetics , Fibroblast Growth Factor 8/genetics , Fibroblast Growth Factor 8/metabolism , Genetic Loci , Genome , Hair Cells, Auditory/ultrastructure , Humans , Multiprotein Complexes/metabolism , Nucleotide Motifs/genetics , Protein Binding , Signal Transduction/genetics
8.
PLoS Genet ; 13(9): e1006967, 2017 Sep.
Article En | MEDLINE | ID: mdl-28892484

The organ of Corti in the cochlea is a two-cell layered epithelium: one cell layer of mechanosensory hair cells that align into one row of inner and three rows of outer hair cells interdigitated with one cell layer of underlying supporting cells along the entire length of the cochlear spiral. These two types of epithelial cells are derived from common precursors in the four- to five-cell layered primordium and acquire functionally important shapes during terminal differentiation through the thinning process and convergent extension. Here, we have examined the role of Six1 in the establishment of the auditory sensory epithelium. Our data show that prior to terminal differentiation of the precursor cells, deletion of Six1 leads to formation of only a few hair cells and defective patterning of the sensory epithelium. Previous studies have suggested that downregulation of Sox2 expression in differentiating hair cells must occur after Atoh1 mRNA activation in order to allow Atoh1 protein accumulation due to antagonistic effects between Atoh1 and Sox2. Our analysis indicates that downregulation of Sox2 in the differentiating hair cells depends on Six1 activity. Furthermore, we found that Six1 is required for the maintenance of Fgf8 expression and dynamic distribution of N-cadherin and E-cadherin in the organ of Corti during differentiation. Together, our analyses uncover essential roles of Six1 in hair cell differentiation and formation of the organ of Corti in the mammalian cochlea.


Cell Differentiation/genetics , Hair Cells, Auditory/metabolism , Homeodomain Proteins/genetics , Organ of Corti/growth & development , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cadherins/genetics , Cochlea/growth & development , Cochlea/metabolism , Epithelium/growth & development , Epithelium/metabolism , Fibroblast Growth Factor 8/genetics , Gene Expression Regulation, Developmental , Homeodomain Proteins/biosynthesis , Mice , Morphogenesis/genetics , Organ of Corti/metabolism , SOXB1 Transcription Factors/genetics
9.
Nat Commun ; 8: 15046, 2017 05 11.
Article En | MEDLINE | ID: mdl-28492243

The adult mammalian cochlear sensory epithelium houses two major types of cells, mechanosensory hair cells and underlying supporting cells, and lacks regenerative capacity. Recent evidence indicates that a subset of supporting cells can spontaneously regenerate hair cells after ablation only within the first week postparturition. Here in vivo clonal analysis of mouse inner ear cells during development demonstrates clonal relationship between hair and supporting cells in sensory organs. We report the identification in mouse of a previously unknown population of multipotent stem/progenitor cells that are capable of not only contributing to the hair and supporting cells but also to other cell types, including glia, in cochlea undergoing development, maturation and repair in response to damage. These multipotent progenitors originate from Eya1-expressing otic progenitors. Our findings also provide evidence for detectable regenerative potential in the postnatal cochlea beyond 1 week of age.


Hair Cells, Auditory/cytology , Hearing/physiology , Labyrinth Supporting Cells/cytology , Multipotent Stem Cells/cytology , Neuroglia/cytology , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Differentiation , Embryo, Mammalian , Female , Gene Expression , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hair Cells, Auditory/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Labyrinth Supporting Cells/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Multipotent Stem Cells/metabolism , Myosin VIIa , Myosins/genetics , Myosins/metabolism , Neuroglia/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Red Fluorescent Protein
10.
Dev Dyn ; 244(7): 866-73, 2015 Jul.
Article En | MEDLINE | ID: mdl-25903664

BACKGROUND: Specification of the metanephric mesenchyme is a central step of kidney development as this mesenchyme promotes nephric duct induction to form a ureteric bud near its caudal end. Before ureteric bud formation, the caudal nephric duct swells to form a pseudostratified epithelial domain that later emerges as the tip of the bud. However, the signals that promote the formation of the transient epithelial domain remain unclear. Here, we investigated the early roles of the mesenchymal factor Six family and its cofactor Eya on the initial induction of nephric duct development. RESULTS: The nephrogenic progenitor population is initially present but significantly reduced in mice lacking both Six1 and Six4 and undertakes an abnormal cell death pathway to be completely eliminated by ∼E10.5-E11.0, similar to that observed in Eya1(-/-) embryos. Consequently, the nephric duct fails to be induced to undergo normal proliferation to pseudostratify and form the ureteric bud in Six1(-/-) ;Six4(-/-) or Eya1(-/-) embryos. CONCLUSIONS: Our data support a model where Eya-Six may form a complex to regulate nephron progenitor cell development before metanephric specification and are critical mesenchymal factors for inducing nephric duct development.


Cell Proliferation/physiology , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nephrons/embryology , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , Stem Cells/metabolism , Trans-Activators/metabolism , Ureter/embryology , Animals , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Models, Biological , Nephrons/cytology , Nuclear Proteins/genetics , Protein Tyrosine Phosphatases/genetics , Stem Cells/cytology , Trans-Activators/genetics , Ureter/cytology
11.
Dev Cell ; 31(4): 434-47, 2014 Nov 24.
Article En | MEDLINE | ID: mdl-25458011

Self-renewal and proliferation of nephron progenitor cells and the decision to initiate nephrogenesis are crucial events directing kidney development. Despite recent advancements in defining lineage and regulators for the progenitors, fundamental questions about mechanisms driving expansion of the progenitors remain unanswered. Here we show that Eya1 interacts with Six2 and Myc to control self-renewing cell activity. Cell fate tracing reveals a developmental restriction of the Eya1(+) population within the intermediate mesoderm to nephron-forming cell fates and a common origin shared between caudal mesonephric and metanephric nephrons. Conditional inactivation of Eya1 leads to loss of Six2 expression and premature epithelialization of the progenitors. Six2 mediates translocation of Eya1 to the nucleus, where Eya1 uses its threonine phosphatase activity to control Myc phosphorylation/dephosphorylation and function in the progenitor cells. Our results reveal a functional link between Eya1, Six2, and Myc in driving the expansion and maintenance of the multipotent progenitors during nephrogenesis.


Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nephrons/cytology , Nephrons/metabolism , Nuclear Proteins/metabolism , Organogenesis/physiology , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Stem Cells/cytology , Transcription Factors/metabolism , Animals , Cell Differentiation/physiology , Mesoderm/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Stem Cells/metabolism
12.
PLoS One ; 9(8): e104503, 2014.
Article En | MEDLINE | ID: mdl-25127126

Proper formation of ureteral smooth muscle cells (SMCs) during embryogenesis is essential for ureter peristalsis that propels urine from the kidney to the bladder in mammals. Currently the molecular factors that regulate differentiation of ureteral mesenchymal cells into SMCs are incompletely understood. A recent study has reported that Smad4 deficiency reduces the number of ureteral SMCs. However, its precise role in the ureteral smooth muscle development remains largely unknown. Here, we used Tbx18:Cre knock-in mouse line to delete Smad4 to examine its requirement in the development of ureteral mesenchyme and SMC differentiation. We found that mice with specific deletion of Smad4 in Tbx18-expressing ureteral mesenchyme exhibited hydroureter and hydronephrosis at embryonic day (E) 16.5, and the mutant mesenchymal cells failed to differentiate into SMCs with increased apoptosis and decreased proliferation. Molecular markers for SMCs including alpha smooth muscle actin (α-SMA) and smooth muscle myosin heavy chain (SM-MHC) were absent in the mutant ureters. Moreover, disruption of Smad4 significantly reduced the expression of genes, including Sox9, Tbx18 and Myocardin associated with SMC differentiation. These findings suggest that Smad4 is essential for initiating the SMC differentiation program during ureter development.


Myocytes, Smooth Muscle/cytology , Organogenesis/genetics , Smad4 Protein/genetics , Ureter/cytology , Actins/metabolism , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Embryonic Development/genetics , Gene Knock-In Techniques , Hydronephrosis/genetics , In Situ Hybridization , Mesoderm/cytology , Mice , Mice, Transgenic , Myosin Heavy Chains/metabolism , Nuclear Proteins/biosynthesis , SOX9 Transcription Factor/biosynthesis , Smooth Muscle Myosins/metabolism , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Trans-Activators/biosynthesis , Ureter/physiology
13.
Dev Biol ; 391(1): 17-31, 2014 Jul 01.
Article En | MEDLINE | ID: mdl-24727670

Tbx18 has been shown to be essential for ureteral development. However, it remains unclear whether it plays a direct role in kidney development. Here we addressed this by focusing on examining the pattern and contribution of Tbx18+ cells in the kidney and its role in kidney vascular development. Expression studies and genetic lineage tracing revealed that Tbx18 is expressed in renal capsule, vascular smooth muscle cells and pericytes and glomerular mesangial cells in the kidney and that Tbx18-expressing progenitors contribute to these cell types. Examination of Tbx18(-/-) kidneys revealed large reduction in vasculature density and dilation of glomerular capillary loops. While SMA+ cells were reduced in the mutant, PDGFRß+ cells were seen in early capillary loop renal corpuscles in the mutant, but fewer than in the controls, and further development of the mesangium failed. Analysis of kidney explants cultured from E12.5 excluded the possibility that the defects observed in the mutant were caused by ureter obstruction. Reduced proliferation in glomerular tuft and increased apoptosis in perivascular mesenchyme were observed in Tbx18(-/-) kidneys. Thus, our analyses have identified a novel role of Tbx18 in kidney vasculature development.


Gene Expression Regulation, Developmental , Glomerular Mesangium/embryology , Kidney/blood supply , Kidney/embryology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/physiology , Animals , Apoptosis , Cell Differentiation , Cell Lineage , Cell Proliferation , Mice , Mice, Transgenic , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Pericytes/metabolism , Receptor, Platelet-Derived Growth Factor beta , Signal Transduction , Time Factors
14.
Development ; 139(11): 1965-77, 2012 Jun.
Article En | MEDLINE | ID: mdl-22513373

Inner ear neurogenesis depends upon the function of the proneural basic helix-loop-helix (bHLH) transcription factors NEUROG1 and NEUROD1. However, the transcriptional regulation of these factors is unknown. Here, using loss- and gain-of-function models, we show that EYA1 and SIX1 are crucial otic neuronal determination factors upstream of NEUROG1 and NEUROD1. Overexpression of both Eya1 and Six1 is sufficient to convert non-neuronal epithelial cells within the otocyst and cochlea as well as the 3T3 fibroblast cells into neurons. Strikingly, all the ectopic neurons express not only Neurog1 and Neurod1 but also mature neuronal markers such as neurofilament, indicating that Eya1 and Six1 function upstream of, and in the same pathway as, Neurog1 and Neurod1 to not only induce neuronal fate but also regulate their differentiation. We demonstrate that EYA1 and SIX1 interact directly with the SWI/SNF chromatin-remodeling subunits BRG1 and BAF170 to drive neurogenesis cooperatively in 3T3 cells and cochlear nonsensory epithelial cells, and that SOX2 cooperates with these factors to mediate neuronal differentiation. Importantly, we show that the ATPase BRG1 activity is required for not only EYA1- and SIX1-induced ectopic neurogenesis but also normal neurogenesis in the otocyst. These findings indicate that EYA1 and SIX1 are key transcription factors in initiating the neuronal developmental program, probably by recruiting and interacting with the SWI/SNF chromatin-remodeling complex to specifically mediate Neurog1 and Neurod1 transcription.


Ear, Inner/innervation , Gene Expression Regulation, Developmental/physiology , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neurogenesis/physiology , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA Primers/genetics , Ear, Inner/embryology , Electroporation , Galactosides , Histological Techniques , Immunohistochemistry , In Situ Hybridization , Indoles , Mice , Mice, Inbred Strains , NIH 3T3 Cells , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Real-Time Polymerase Chain Reaction , Two-Hybrid System Techniques
15.
Dev Cell ; 22(2): 377-90, 2012 Feb 14.
Article En | MEDLINE | ID: mdl-22340499

Inner-ear hair cell differentiation requires Atoh1 function, while Eya1, Six1, and Sox2 are coexpressed in sensory progenitors and mutations in these genes cause sensorineural hearing loss. However, how these genes are linked functionally and the transcriptional networks controlling hair cell induction remain unclear. Here, we show (1) that Eya1/Six1 are necessary for hair cell development, and their coexpression in mouse cochlear explants is sufficient to induce hair cell fate in the nonsensory epithelium expressing low-level Sox2 by activating not only Atoh1-dependent but also Atoh1-independent pathways and (2) that both pathways induce Pou4f3 to promote hair cell differentiation. Sox2 cooperates with Eya1/Six1 to synergistically activate Atoh1 transcription via direct binding to the conserved Sox- and Six-binding sites in Atoh1 enhancers, and these proteins physically interact. Our findings demonstrate that direct and cooperative interactions between the Sox2, Six1, and Eya1 proteins coordinate Atoh1 expression to specify hair cell fate.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Cochlea/metabolism , Hair Cells, Auditory/metabolism , Homeodomain Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Blotting, Western , Cell Differentiation , Chromatin Immunoprecipitation , Cochlea/cytology , Electrophoretic Mobility Shift Assay , Electroporation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Immunoenzyme Techniques , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mutation/genetics , Nuclear Proteins/genetics , Phosphorylation , Protein Tyrosine Phosphatases/genetics , SOXB1 Transcription Factors/genetics
16.
Dev Biol ; 352(1): 141-51, 2011 Apr 01.
Article En | MEDLINE | ID: mdl-21281623

Urinary tract morphogenesis requires subdivision of the ureteric bud (UB) into the intra-renal collecting system and the extra-renal ureter, by responding to signals in its surrounding mesenchyme. BMP4 is a mesenchymal regulator promoting ureter development, while GREM1 is necessary to negatively regulate BMP4 activity to induce UB branching. However, the mechanisms that regulate the GREM1-BMP4 signaling are unknown. Previous studies have shown that Six1-deficient mice lack kidneys, but form ureters. Here, we show that the tip cells of Six1(-/-) UB fail to form an ampulla for branching. Instead, the UB elongates within Tbx18- and Bmp4-expressing mesenchyme. We find that the expression of Grem1 in the metanephric mesenchyme (MM) is Six1-dependent. Treatment of Six1(-/-) kidney rudiments with GREM1 protein restores ampulla formation and branching morphogenesis. Furthermore, we demonstrate that genetic reduction of BMP4 levels in Six1(-/-) (Six1(-/-); Bmp4(+/-)) embryos restores urinary tract morphogenesis and kidney formation. This study uncovers an essential function for Six1 in the MM as an upstream regulator of Grem1 in initiating branching morphogenesis.


Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Mesoderm/embryology , Morphogenesis , Animals , Bone Morphogenetic Protein 4/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Gene Dosage/drug effects , Gene Expression Regulation, Developmental/drug effects , Gene Silencing/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney/embryology , Kidney/metabolism , Mesoderm/cytology , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Morphogenesis/drug effects , Organogenesis/drug effects , Recombinant Proteins/pharmacology , T-Box Domain Proteins/metabolism , Up-Regulation/drug effects , Ureter/cytology , Ureter/drug effects , Ureter/embryology , Ureter/metabolism , Wnt Proteins/metabolism
17.
Immunology ; 128(1 Suppl): e506-13, 2009 Sep.
Article En | MEDLINE | ID: mdl-19740311

In our previous study, the hinge fragment (225-232/225'-232') of human immunoglobulin G1 (IgG1) was used as a space peptide linker for synthesizing the GnRH3-hinge-MVP chimeric peptide, whereby three repeated gonadotrophin-releasing hormone (GnRH) units and a T-cell epitope from measles virus fusion protein (MVP) were amide-bond-linked at the N and C terminus, respectively, to the hinge peptide for producing anti-GnRH antibody responses. To investigate whether or not the hinge region fragment can improve the immunogenicity of GnRH, we further synthesized and purified GnRH3-hinge-MVP, GnRH3-hinge and GnRH3-MVP using recombinant DNA technology. Under high pH conditions, GnRH3-hinge-MVP was capable of forming double-chain structures. Immunization of male mice with the immunogens of GnRH3-hinge-MVP resulted in the generation of high-titre antibodies specific for GnRH. The synthetic GnRH3-hinge and GnRH3-MVP induced a lower titre of anti-GnRH antibody than GnRH3-hinge-MVP. This was followed by a decrease in serum testosterone levels, which resulted in a low level of expression of the relaxin-like factor gene in the testis. Our data suggest that peptide and T-cell epitopes oriented at the N-terminus or C-terminus of hinge peptides simplify the antigenic peptide conjugates and may be considered as potential synthetic immunogens.


Epitopes, T-Lymphocyte/immunology , Gonadotropin-Releasing Hormone/immunology , Vaccines, Subunit/immunology , Viral Proteins/immunology , Animals , Antibodies/blood , Gene Expression/immunology , Gonadotropin-Releasing Hormone/metabolism , Guanine Nucleotide Exchange Factors , Immunoglobulin G/immunology , Insulin/immunology , Insulin/metabolism , Male , Mice , Oxidation-Reduction , Proteins/immunology , Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Testis/immunology , Testis/metabolism , Testosterone/blood , Transcription Factors/immunology , Transcription Factors/metabolism , Viral Proteins/metabolism
18.
J Neurochem ; 106(4): 1900-13, 2008 Aug.
Article En | MEDLINE | ID: mdl-18624924

Several lines of evidence suggest involvement of NMDA receptors (NMDARs) in the regulation of neurogenesis in adults and the formation of spatial memory. Functional properties of NMDARs are strongly influenced by the type of NR2 subunits incorporated. In adult forebrain regions such as the hippocampus and cortex, only NR2A and NR2B subunits are available to form the receptor complex with NR1 subunit. NR2B is predominant NR2 subunit in any of rat or human neural stem cells (NSCs). Thus, we suppose that NR2B-containing NMDAR should be critical in regulating adult neurogenesis, and thereby playing a role in the formation of spatial memory. In the cultured NSCs derived from the embryonic brain of rats, NR2B subunit-specific NMDAR antagonist Ro25-6981 increased cell proliferation, whereas MK-801, non-selective open-channel blocker of NMDARs, inhibited cell proliferation. Blockade of NR2B-containing NMDAR stimulated neurogenesis in the adult hippocampus and facilitated the formation of spatial memory. The enhanced spatial memory dropped back to base level when the NR2B antagonist-induced neurogenesis was neutralized by 3'-azido-deoxythymidine, a telomerase inhibitor. In addition, blockade of NR2B inhibited neuronal nitric oxide synthase (nNOS) enzymatic activity. In null mutant mice lacking nNOS gene (nNOS-/-), the effects of NR2B antagonist on neurogenesis disappeared. Moreover, nitric oxide donor DETA/NONOate attenuated and nNOS inhibitor 7-nitroindazole enhanced the effect of Ro 25-6981 on NSCs proliferation. Our findings suggest that NR2B-containing NMDAR subtypes negatively regulate neurogenesis in the adult hippocampus by activating nNOS activity and thereby hinder the formation of spatial memory.


Down-Regulation/physiology , Hippocampus/physiology , Memory/physiology , Neural Inhibition/physiology , Neurons/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Spatial Behavior/physiology , Animals , Animals, Newborn , Cells, Cultured , Hippocampus/cytology , Hippocampus/metabolism , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Nitric Oxide Synthase Type I/physiology , Rats , Rats, Sprague-Dawley
19.
Cancer Lett ; 259(2): 240-50, 2008 Feb 08.
Article En | MEDLINE | ID: mdl-18039558

We have previously shown that anti-GnRH antibodies responses can be induced by synthetic GnRH3-hinge-MVP peptide. In this study, GnRH3-hinge-MVP of conjugation to heat shock protein 65 was used as an adjuvant-free vaccine to assess the therapeutic effects of GnRH immunoneutralisation on tumor development in the mice model. Compared with mice treated with Hsp65 and PBS, mice of the o.t. model receiving in situ treatment GnRH3-hinge-MVP-Hsp65 had significant prolongation of survival and suppression of local tumor growth. Serum levels of both testosterone and luteinizing hormone were reduced by treatment with GnRH3-hinge-MVP-Hsp65 (p<0.05). Further analyses of cell mediated immune responses showed that GnRH3-hinge-MVP-Hsp65 induced stronger lymphocyte proliferative responses and higher levels of IFN-gamma (p<0.001). The conjugation of the recombinant GnRH peptide to Hsp65 could be considered a promising approach for the development of an efficacious vaccine against the prostate cancer.


Antibody Formation , Bacterial Proteins/immunology , Cancer Vaccines/immunology , Chaperonins/immunology , Gonadotropin-Releasing Hormone/immunology , Immunity, Cellular , Prostatic Neoplasms/prevention & control , Viral Proteins/immunology , Animals , Cell Line, Tumor , Cell Proliferation , Chaperonin 60 , Immunoglobulin G/blood , Interferon-gamma/metabolism , Luteinizing Hormone/blood , Lymphocyte Activation , Lymphocytes/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Prostatic Neoplasms/blood , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Recombinant Proteins/immunology , Testosterone/blood , Time Factors , Vaccines, Conjugate/immunology , Vaccines, Synthetic/immunology
20.
J Neurochem ; 103(5): 1843-54, 2007 Dec.
Article En | MEDLINE | ID: mdl-17854383

Increasing evidence suggests that depression may be associated with a lack of hippocampal neurogenesis. It is well established that neuronal nitric oxide synthase (nNOS)-derived NO exerts a negative control on the hippocampal neurogenesis. Using genetic and pharmacological methods, we investigated the roles of nNOS in depression induced by chronic mild stress (CMS) in mice. Hippocampal nNOS over-expression was first observed 4 days and remained elevated 21 and 56 days after exposure to CMS. The mice exposed to CMS exhibited behavioral changes typical of depression, and impaired neurogenesis in the hippocampus. The CMS-induced behavioral despair and hippocampal neurogenesis impairment were prevented and reversed in the null mutant mice lacking nNOS gene (nNOS-/-) and in the mice receiving nNOS inhibitor. Disrupting hippocampal neurogenesis blocked the antidepressant effect of nNOS inhibition. Moreover, nNOS-/- mice exhibited antidepressant-like properties. Our findings suggest that nNOS over-expression in the hippocampus is essential for chronic stress-induced depression and inhibiting nNOS signaling in brain may represent a novel approach for the treatment of depressive disorders.


Cell Proliferation , Depression/etiology , Hippocampus/pathology , Neurons/physiology , Nitric Oxide Synthase Type I/physiology , Stress, Psychological/complications , Analysis of Variance , Animals , Behavior, Animal , Bromodeoxyuridine , Depression/pathology , Drug Interactions , Enzyme Inhibitors/pharmacology , Hindlimb Suspension/methods , Indazoles/pharmacology , Male , Mice , Mice, Knockout , Nitric Oxide Synthase Type I/deficiency , Time Factors , Zidovudine/pharmacology
...