Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Dev Biol ; 429(1): 356-369, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28449850

ABSTRACT

The migration and fate of cranial and vagal neural crest-derived progenitor cells (NCPCs) have been extensively studied; however, much less is known about sacral NCPCs particularly in regard to their distribution in the urogenital system. To construct a spatiotemporal map of NCPC migration pathways into the developing lower urinary tract, we utilized the Sox10-H2BVenus transgene to visualize NCPCs expressing Sox10. Our aim was to define the relationship of Sox10-expressing NCPCs relative to bladder innervation, smooth muscle differentiation, and vascularization through fetal development into adulthood. Sacral NCPC migration is a highly regimented, specifically timed process, with several potential regulatory mileposts. Neuronal differentiation occurs concomitantly with sacral NCPC migration, and neuronal cell bodies are present even before the pelvic ganglia coalesce. Sacral NCPCs reside within the pelvic ganglia anlagen through 13.5 days post coitum (dpc), after which they begin streaming into the bladder body in progressive waves. Smooth muscle differentiation and vascularization of the bladder initiate prior to innervation and appear to be independent processes. In adult bladder, the majority of Sox10+ cells express the glial marker S100ß, consistent with Sox10 being a glial marker in other tissues. However, rare Sox10+ NCPCs are seen in close proximity to blood vessels and not all are S100ß+, suggesting either glial heterogeneity or a potential nonglial role for Sox10+ cells along vasculature. Taken together, the developmental atlas of Sox10+ NCPC migration and distribution profile of these cells in adult bladder provided here will serve as a roadmap for future investigation in mouse models of lower urinary tract dysfunction.


Subject(s)
Cell Movement , Neural Crest/cytology , Sacrum/cytology , Urogenital System/innervation , Animals , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Endothelium, Vascular/metabolism , Ganglia/metabolism , Mesoderm/metabolism , Mice, Transgenic , Myocytes, Smooth Muscle/cytology , Neural Crest/metabolism , Neuroglia/cytology , Neuroglia/metabolism , SOXE Transcription Factors/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Time Factors , Urogenital System/blood supply
2.
J Cell Sci ; 125(Pt 9): 2141-7, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22357946

ABSTRACT

Planar cell polarity (PCP) describes the polarized orientation of cells within the plane of a tissue. Unlike epithelial PCP, the mechanisms underlying PCP signaling in migrating cells remain undefined. Here, the establishment of PCP must be coordinated with dynamic changes in cell adhesion and extracellular matrix (ECM) organization. During gastrulation, the membrane type-1 matrix metalloproteinase (MT1-MMP or MMP14) is required for PCP and convergence and extension cell movements. We report that the PCP protein Vang-like 2 (VANGL2) regulates the endocytosis and cell-surface availability of MMP14 in manner that is dependent on focal adhesion kinase. We demonstrate that zebrafish trilobite/vangl2 mutant embryos exhibit increased Mmp14 activity and decreased ECM. Furthermore, in vivo knockdown of Mmp14 partially rescues the Vangl2 loss-of-function convergence and extension phenotype. This study identifies a mechanism linking VANGL2 with MMP14 trafficking and suggests that establishment of PCP in migrating gastrula cells requires regulated proteolytic degradation or remodeling of the ECM. Our findings implicate matrix metalloproteinases as downstream effectors of PCP and suggest a broadly applicable mechanism whereby VANGL2 affects diverse morphogenetic processes.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gastrula/metabolism , Gastrulation/genetics , Matrix Metalloproteinase 14/metabolism , Membrane Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Adhesion/physiology , Cell Membrane/metabolism , Cell Movement/physiology , Cell Polarity/physiology , Embryo, Nonmammalian , Endocytosis/physiology , Extracellular Matrix/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gastrula/embryology , Gene Knockdown Techniques , Matrix Metalloproteinase 14/genetics , Membrane Proteins/genetics , Mutation , Protein Transport/physiology , Proteolysis , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
3.
Genesis ; 49(7): 599-618, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21504042

ABSTRACT

To facilitate dynamic imaging of neural crest (NC) lineages and discrimination of individual cells in the enteric nervous system (ENS) where close juxtaposition often complicates viewing, we generated a mouse BAC transgenic line that drives a Histone2BVenus (H2BVenus) reporter from Sox10 regulatory regions. This strategy does not alter the endogenous Sox10 locus and thus facilitates analysis of normal NC development. Our Sox10-H2BVenus BAC transgene exhibits temporal, spatial, and cell-type specific expression that reflects endogenous Sox10 patterns. Individual cells exhibiting nuclear-localized fluorescence of the H2BVenus reporter are readily visualized in both fixed and living tissue and are amenable to isolation by fluorescence activated cell sorting (FACS). FACS-isolated H2BVenus+ enteric NC-derived progenitors (ENPs) exhibit multipotency, readily form neurospheres, self-renew in vitro and express a variety of stem cell genes. Dynamic live imaging as H2BVenus+ ENPs migrate down the fetal gut reveals cell fragmentation suggesting that apoptosis occurs at a low frequency during normal development of the ENS. Confocal imaging both during population of the fetal intestine and in postnatal gut muscle strips revealed differential expression between individual cells consistent with down-regulation of the transgene as progression towards non-glial fates occurs. The expression of the Sox10-H2BVenus transgene in multiple regions of the peripheral nervous system will facilitate future studies of NC lineage segregation as this tool is expressed in early NC progenitors and maintained in enteric glia.


Subject(s)
Enteric Nervous System , Gene Expression Regulation, Developmental , Histones/genetics , Molecular Imaging , SOXE Transcription Factors/genetics , Stem Cells , Transgenes/genetics , Alleles , Animals , Cell Movement/genetics , Cell Proliferation , Cells, Cultured , Enteric Nervous System/embryology , Enteric Nervous System/metabolism , Female , Gene Order , Histones/metabolism , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neural Crest/embryology , Neural Crest/metabolism , Organ Specificity/genetics , Stem Cells/cytology , Stem Cells/metabolism
4.
Hum Mol Genet ; 19(22): 4353-72, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20739296

ABSTRACT

Abnormalities in the development of enteric neural crest-derived progenitors (ENPs) that generate the enteric nervous system (ENS) can lead to aganglionosis in a variable portion of the distal gastrointestinal tract. Cumulative evidence suggests that variation of aganglionosis is due to gene interactions that modulate the ability of ENPs to populate the intestine; however, the developmental processes underlying this effect are unknown. We hypothesized that differences in enteric ganglion deficits could be attributable to the effects of genetic background on early developmental processes, including migration, proliferation, or lineage divergence. Developmental processes were investigated in congenic Sox10(Dom) mice, an established Hirschsprung disease (HSCR) model, on distinct inbred backgrounds, C57BL/6J (B6) and C3HeB/FeJ (C3Fe). Immuno-staining on whole-mount fetal gut tissue and dissociated cell suspensions was used to assess migration and proliferation. Flow cytometry utilizing the cell surface markers p75 and HNK-1 was used to isolate live ENPs for analysis of developmental potential. Frequency of ENPs was reduced in Sox10(Dom) embryos relative to wild-type embryos, but was unaffected by genetic background. Both migration and developmental potential of ENPs in Sox10(Dom) embryos were altered by inbred strain background with the most highly significant differences seen for developmental potential between strains and genotypes. In vivo imaging of fetal ENPs and postnatal ganglia demonstrates that altered lineage divergence impacts ganglia in the proximal intestine. Our analysis demonstrates that genetic background alters early ENS development and suggests that abnormalities in lineage diversification can shift the proportions of ENP populations and thus may contribute to ENS deficiencies in vivo.


Subject(s)
Enteric Nervous System/embryology , Hirschsprung Disease/genetics , Neural Crest/cytology , SOXE Transcription Factors/genetics , Stem Cells/cytology , Animals , CD57 Antigens/metabolism , Disease Models, Animal , Enteric Nervous System/cytology , Enteric Nervous System/metabolism , Ganglia/embryology , Ganglia/pathology , Hirschsprung Disease/embryology , Hirschsprung Disease/metabolism , Humans , Immunohistochemistry , Intestine, Small/metabolism , Intestine, Small/pathology , Intestines/cytology , Intestines/embryology , Mice , Mice, Congenic , Mice, Inbred C3H , Mice, Inbred C57BL , Mutation , Neural Crest/embryology , Species Specificity
5.
Cancer Lett ; 287(1): 54-61, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19577357

ABSTRACT

Van Gogh-Like 2 (VANGL2) is a planar cell polarity protein essential for collective migration during embryonic development, yet its contribution to tumor cell motility and invasion are unknown. We report for the first time that loss of VANGL2 in human cancer cells promotes efficient collective and directed migration and matrix metalloproteinase (MMP)-dependent ECM invasion. We show that VANGL2 knockdown cells exhibit increased activation of secreted MMP2, higher levels of membrane-localized MMP14, and decreased cell-surface fibronectin. These important findings support the notion that planar cell polarity proteins act in coordination with known regulators of cancer cell migration to influence invasion and perhaps metastasis.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Matrix Metalloproteinase 14/physiology , Matrix Metalloproteinase 2/physiology , Membrane Proteins/physiology , Neoplasm Invasiveness , Cell Line, Tumor , Cell Movement , Cell Polarity , Humans
6.
Dev Dyn ; 237(4): 1119-32, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18351668

ABSTRACT

The mammalian enteric nervous system (ENS) derives from migratory enteric neural crest-derived cells (ENCC) that express the transcription factor Phox2b. Studies of these enteric progenitors have typically relied on immunohistochemical (IHC) detection. To circumvent complicating factors of IHC, we have generated a mouse BAC transgenic line that drives a Histone2BCerulean (H2BCFP) reporter from Phox2b regulatory regions. This construct does not alter the endogenous Phox2b locus and enables studies of normal neural crest (NC) derivatives. The Phox2b-H2BCFP transgene expresses the H2BCFP reporter in patterns that recapitulate expression of endogenous Phox2b. Our studies reveal Phox2b expression in mature enteric glia at levels below that of enteric neurons. Moreover, we also observe differential expression of the transgene reporter within the leading ENCC that traverse the gut. Our findings indicate that the wavefront of migrating enteric progenitors is not homogeneous, and suggest these cells may be fate-specified before expression of mature lineage markers appears.


Subject(s)
Enteric Nervous System , Histones/metabolism , Homeodomain Proteins/metabolism , Neural Crest , Neuroglia/physiology , Transcription Factors/metabolism , Transgenes , Animals , Cell Movement/physiology , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Bacterial/metabolism , Enteric Nervous System/anatomy & histology , Enteric Nervous System/embryology , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Histones/genetics , Homeodomain Proteins/genetics , Intestinal Mucosa/metabolism , Intestines/embryology , Intestines/innervation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Crest/cytology , Neural Crest/physiology , Neuroglia/cytology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/genetics
7.
Dev Dyn ; 235(5): 1413-32, 2006 May.
Article in English | MEDLINE | ID: mdl-16586440

ABSTRACT

Sox10 is an essential transcription factor required for development of neural crest-derived melanocytes, peripheral glia, and enteric ganglia. Multiple transcriptional targets regulated by Sox10 have been identified; however, little is known regarding regulation of Sox10. High sequence conservation surrounding 5' exons 1 through 3 suggests these regions might contain functional regulatory elements. However, we observed that these Sox10 genomic sequences do not confer appropriate cell-specific transcription in vitro when linked to a heterologous reporter. To identify elements required for expression of Sox10 in vivo, we modified bacterial artificial chromosomes (BACs) to generate a Sox10betaGeoBAC transgene. Our approach leaves endogenous Sox10 loci unaltered, circumventing haploinsufficiency issues that arise from gene targeting. Sox10betaGeoBAC expression closely approximates Sox10 expression in vivo, resulting in expression in anterior dorsal neural tube at embryonic day (E) 8.5 and in cranial ganglia, otic vesicle, and developing dorsal root ganglia at E10.5. Characterization of Sox10betaGeoBAC expression confirms the presence of essential regulatory regions and additionally identifies previously unreported expression in thyroid parafollicular cells, thymus, salivary, adrenal, and lacrimal glands. Fortuitous deletions in independent Sox10betaGeoBAC lines result in loss of transgene expression in peripheral nervous system lineages and coincide with evolutionarily conserved regions. Our analysis indicates that Sox10 expression requires the presence of distant cis-acting regulatory elements. The Sox10betaGeoBAC transgene offers one avenue for specifically testing the role of individual conserved regions in regulation of Sox10 and makes possible analysis of Sox10+ derivatives in the context of normal neural crest development.


Subject(s)
Chromosomes, Artificial, Bacterial , Enteric Nervous System/embryology , Enteric Nervous System/metabolism , High Mobility Group Proteins/biosynthesis , High Mobility Group Proteins/genetics , Neural Crest/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transgenes , 5' Flanking Region/genetics , Animals , Cell Line, Tumor , Female , Male , Melanoma, Experimental , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Neural Crest/embryology , Rats , SOXE Transcription Factors
8.
Hum Mol Genet ; 13(19): 2289-301, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15294878

ABSTRACT

Cumulative evidence suggests that Hirschsprung disease (HSCR) is the consequence of multiple gene interactions that modulate the ability of enteric neural crest (NC) cells to populate the developing gut. One of the essential genes for this process is the NC transcription factor Sox10. Sox10Dom mice on a mixed genetic background show variation in penetrance and expressivity of enteric aganglionosis that are analogous to the variable aganglionosis seen in human HSCR families. The phenotype of Sox10Dom mice in congenic lines indicates this variation arises from modifiers in the genetic background. To determine whether known HSCR susceptibility loci are acting as modifiers of Sox10, we tested for association between genes in the endothelin signaling pathway (EdnrB, Edn3, Ece1) and severity of aganglionosis in an extended pedigree of B6C3FeLe.Sox10Dom mice. Single locus association analysis in this pedigree identifies interaction between EdnrB and Sox10. Additional analysis of F2 intercross progeny confirms a highly significant effect of EdnrB alleles on the Sox10Dom/+ phenotype. The presence of C57BL/6J alleles at EdnrB is associated with increased penetrance and more severe aganglionosis in Sox10Dom mutants. Crosses between EdnrB and Sox10 mutants corroborate this gene interaction with double mutant progeny exhibiting significantly more severe aganglionosis. The background strain of the EdnrB mutant further influences the phenotype of Sox10/EdnrB double mutant progeny implying the action of additional modifiers on this phenotype. Our data demonstrates that Sox10-EdnrB interactions can influence development of the enteric nervous system in mouse models and suggests that this interaction could contribute to the epistatic network producing variation between patients with aganglionosis.


Subject(s)
DNA-Binding Proteins/physiology , Enteric Nervous System/embryology , High Mobility Group Proteins/physiology , Hirschsprung Disease/genetics , Hirschsprung Disease/pathology , Receptor, Endothelin B/metabolism , Animals , Crosses, Genetic , DNA-Binding Proteins/genetics , Endothelins/metabolism , Enteric Nervous System/growth & development , Genes, Dominant , High Mobility Group Proteins/genetics , Hirschsprung Disease/embryology , Hirschsprung Disease/metabolism , Mice , Mice, Congenic , Mice, Inbred C3H , Mice, Inbred C57BL , Mutation , Pedigree , SOXE Transcription Factors , Severity of Illness Index , Signal Transduction , Transcription Factors
9.
Nat Genet ; 36(7): 732-7, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15170213

ABSTRACT

Hirschsprung disease (HSCR) is a multigenic, congenital disorder that affects 1 in 5,000 newborns and is characterized by the absence of neural crest-derived enteric ganglia in the colon. One of the primary genes affected in HSCR encodes the G protein-coupled endothelin receptor-B (EDNRB). The expression of Ednrb is required at a defined time period during the migration of the precursors of the enteric nervous system (ENS) into the colon. In this study, we describe a conserved spatiotemporal ENS enhancer of Ednrb. This 1-kb enhancer is activated as the ENS precursors approach the colon, and partial deletion of this enhancer at the endogenous Ednrb locus results in pigmented mice that die postnatally from megacolon. We identified binding sites for SOX10, an SRY-related transcription factor associated with HSCR, in the Ednrb ENS enhancer, and mutational analyses of these sites suggested that SOX10 may have multiple roles in regulating Ednrb in the ENS.


Subject(s)
Gene Expression Regulation/physiology , High Mobility Group Proteins/physiology , Neoplasm Proteins/physiology , Receptors, Endothelin/genetics , Animals , Base Sequence , Enhancer Elements, Genetic , Enteric Nervous System/physiology , Mice , Mice, Transgenic , Molecular Sequence Data , SOXE Transcription Factors , Sequence Homology, Nucleic Acid , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...