Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Placenta ; 31(9): 747-55, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20659767

ABSTRACT

Colonisation of the maternal uterine wall by the trophoblast involves a series of alterations in the behaviour and morphology of trophoblast cells. Villous cytotrophoblast cells change from a well-organised coherently layered phenotype to one that is extravillous, acquiring a proliferative, migratory and invasive capacity, to facilitate fetal-maternal interaction. These changes are similar to those of other developmental processes falling under the umbrella of an epithelial-mesenchymal transition (EMT). Modulation of cell adhesion and cell polarity occurs through changes in cell-cell junctional molecules, such as the cadherins. The cadherins, particularly the classical cadherins (e.g. Epithelial-(E)-cadherin), and their link to adaptors called catenins at cell-cell contacts, are important for maintaining cell attachment and the layered phenotype of the villous cytotrophoblast. In contrast, reduced expression and re-organization of cadherins from these cell junctional regions promote a loosened connection between cells, coupled with reduced apico-basal polarity. Certain non-classical cadherins play an active role in cell migration processes. In addition to the classical cadherins, two other cadherins which have been reported in placental tissues are vascular endothelial (VE) cadherin and cadherin-11. Cadherin molecules are well placed to be key regulators of trophoblast cell behaviour, analogous to their role in other developmental EMTs. This review addresses cadherin expression and function in normal and diseased human placental tissues, especially in fetal growth restriction and pre-eclampsia where trophoblast invasion is reduced.


Subject(s)
Cadherins/physiology , Epithelial-Mesenchymal Transition/physiology , Placentation/physiology , Cadherins/biosynthesis , Cell Adhesion , Cell Movement/genetics , Female , Fetal Growth Retardation/physiopathology , Humans , Placenta/metabolism , Pre-Eclampsia/physiopathology , Pregnancy , Trophoblasts/cytology
2.
Neurogastroenterol Motil ; 22(3): 331-e86, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19775251

ABSTRACT

BACKGROUND Most enteric neurones arise from neural crest cells that originate in the post-otic hindbrain, and migrate into and along the developing gastrointestinal tract. There is currently great interest in the possibility of cell therapy to replace diseased or absent enteric neurones in patients with enteric neuropathies, such as Hirschsprung's disease. However, it is unclear whether neural crest stem/progenitor cells will be able to colonize colon (i) in which the mesenchyme has differentiated into distinct layers, (ii) that already contains enteric neurones or (iii) that lacks a gene expressed by the gut mesenchyme, such as endothelin-3 (Et-3). METHODS Co-cultures were used to examine the ability of enteric neural crest-derived cells (ENCCs) from E11.5 mouse gut to colonize a variety of recipient hindguts. KEY RESULTS Enteric neural crest-derived cells migrated and gave rise to neurones in E14.5 and E16.5 aneural colon in which the external muscle layers had differentiated, but they did not migrate as far as in younger colon. There was no evidence of altered ENCC proliferation, cell death or neuronal differentiation in older recipient explants. Enteric neural crest-derived cells failed to enter most recipient E14.5 and E16.5 colon explants already containing enteric neurones, and the few that did showed very limited migration. Finally, ENCCs migrated a shorter distance and a higher proportion expressed the pan-neuronal marker, Hu, in recipient E11.5 Et-3(-/-) colon compared to wild-type recipient colon. CONCLUSIONS & INFERENCES Age and an absence of Et-3 from the recipient gut both significantly reduced but did not prevent ENCC migration, but the presence of neurones almost totally prevented ENCC migration.


Subject(s)
Cell Movement/physiology , Colon/innervation , Endothelin-3/metabolism , Neurogenesis/physiology , Neurons/physiology , Age Factors , Analysis of Variance , Animals , Coculture Techniques , Colon/cytology , Colon/metabolism , ELAV Proteins/metabolism , Endothelin-3/genetics , Immunohistochemistry , Mice , Mice, Knockout , Neural Crest/cytology , Neural Crest/physiology , Neurons/cytology , Stem Cells/cytology , Stem Cells/physiology
3.
Dev Dyn ; 238(12): 3152-5, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19877279

ABSTRACT

Gene expression fields in embryogenesis are spatially precise and often small, so experimental gene expression often requires similar spatial definition. For in ovo electroporation, typically a gene construct is injected into a natural body cavity in the embryo prior to electroporation. Limited control of the size and location of the electroporated field can be obtained by varying electrode placement and geometry, and by altering the miscibility and viscosity of the construct vehicle but it is difficult to tightly constrain electroporation to small regions. Electroporation of different constructs in close proximity has not been possible. We show that loading the construct into an agarose bead, which is then microsurgically implanted, allows for focal electroporation. Different constructs can be electroporated in close proximity by emplacing several agarose beads. This technique is simple, cheap, rapid, and requires no more specialised equipment than that required for conventional in ovo electroporation.


Subject(s)
Electroporation/methods , Gene Transfer Techniques , Ovum , Animals , Animals, Genetically Modified , Embryo, Nonmammalian , Fluorescent Antibody Technique/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Microspheres , Ovum/cytology , Quail/embryology , Transgenes
4.
Endocrinology ; 150(9): 4044-55, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19477940

ABSTRACT

We have recently demonstrated that fibroblast growth factor (FGF)-2 promotes neuroblastoma cell differentiation and overrides their mitogenic response to IGF-I. However, the mechanisms involved are unknown. SK-N-MC cells were cultured with FGF-2 (50 ng/ml) and/or IGF-I (100 ng/ml) up to 48 h. Fluorescence-activated cell sorting analysis indicated that FGF-2 promotes G1/G0 cell cycle phase arrest. Gene expression by RT2-PCR and cellular localization showed up-regulation of p21. We then investigated whether FGF-2-induced differentiation of SK-N-MC cells (by GAP43 and NeuroD-6 expression) involves epithelium-mesenchyme transition interconversion. Real-time PCR (RT2-PCR) showed modulation of genes involved in maintenance of the epithelial phenotype and cell-matrix interactions (E-cadherin, Snail-1, MMPs). Zymography confirmed FGF-2 up-regulated MMP2 and induced MMP9, known to contribute to neuronal differentiation and neurite extension. Id1-3 expression was determined by RT2-PCR. FGF-2 induced Id2, while down-regulating Id1 and Id3. FGF-2 induced nuclear accumulation of ID2 protein, while ID1 and ID3 remained cytoplasmic. RNA interference demonstrated that Id3 regulates differentiation and cell cycle (increased Neuro-D6 and p21 mRNA), while d Id2 modulates epithelium-mesenchyme transition-like events (increased E-cadherin mRNA). In conclusion, we have shown for the first time that FGF-2 induces differentiation of neuroblastoma cells via activation of a complex gene expression program enabling modulation of cell cycle, transcription factors, and suppression of the cancer phenotype. The use of RNA interference indicated that Id-3 is a key regulator of these events, thus pointing to a novel therapeutic target for this devastating childhood cancer.


Subject(s)
Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p21/physiology , Fibroblast Growth Factor 2/physiology , G1 Phase/drug effects , Inhibitor of Differentiation Protein 1/physiology , Inhibitor of Differentiation Protein 2/physiology , Inhibitor of Differentiation Proteins/physiology , Neoplasm Proteins/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Extracellular Matrix/physiology , Gene Expression Regulation, Neoplastic , Humans , Neuroblastoma/genetics , RNA Interference , RNA, Messenger/metabolism , Signal Transduction
5.
Dev Biol ; 270(2): 455-73, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15183726

ABSTRACT

Neural crest-derived cells that form the enteric nervous system undergo an extensive migration from the caudal hindbrain to colonize the entire gastrointestinal tract. Mice in which the expression of GFP is under the control of the Ret promoter were used to visualize neural crest-derived cell migration in the embryonic mouse gut in organ culture. Time-lapse imaging revealed that GFP(+) crest-derived cells formed chains that displayed complicated patterns of migration, with sudden and frequent changes in migratory speed and trajectories. Some of the leading cells and their processes formed a scaffold along which later cells migrated. To examine the effect of population size on migratory behavior, a small number of the most caudal GFP(+) cells were isolated from the remainder of the population. The isolated cells migrated slower than cells in large control populations, suggesting that migratory behavior is influenced by cell number and cell-cell contact. Previous studies have shown that neurons differentiate among the migrating cell population, but it is unclear whether they migrate. The phenotype of migrating cells was examined. Migrating cells expressed the neural crest cell marker, Sox10, but not neuronal markers, indicating that the majority of migratory cells observed did not have a neuronal phenotype.


Subject(s)
Cell Movement/physiology , Gastrointestinal Tract/cytology , Mice/embryology , Neural Crest/embryology , Animals , Cell Count , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Female , Gastrointestinal Tract/physiology , Green Fluorescent Proteins , High Mobility Group Proteins/metabolism , Immunohistochemistry , Luminescent Proteins/metabolism , Mice/metabolism , Mice, Transgenic , Microscopy, Confocal , Pregnancy , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/metabolism , SOXE Transcription Factors , Time Factors , Transcription Factors
6.
J Cell Physiol ; 199(3): 371-80, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15095284

ABSTRACT

The insulin-like growth factor (IGF) system is a key regulator of cell growth, survival and differentiation, and these functions are co-modulated by other growth factors including fibroblast growth factor-2 (FGF-2). To investigate IGF/FGF interactions in neuronal cells, we employed neuroblastoma cells (SK-N-MC). In serum free conditions proliferation of the SK-N-MC cells was promoted by IGF-I (25 ng/ml), but blunted by FGF-2 (50 ng/ml). IGF-I-induced proliferation was abolished in the presence of FGF-2 even when IGF-I was used at 100 ng/ml. In addition to our previously described FGF-2 induced proteolytic cleavage of IGFBP-2, we found that FGF-2 increased IGFBP-6 levels in conditioned medium (CM) without affecting IGFBP-6 mRNA abundance. Modulation of IGFBP-2 and -6 levels were not significant mechanisms involved in the blockade of IGF-I action since the potent IGF-I analogues [QAYL]IGF-I and des(1-3)IGF-I (minimal IGFBP affinity) were unable to overcome FGF-2 inhibition of cell proliferation. FGF-2 treated cells showed morphological differentiation expressing the TUJ1 neuronal marker while cells treated with IGF-I alone showed no morphological change. When IGF-I was combined with FGF-2, however, cell morphology was indistinguishable from that seen with FGF-2 alone. FGF-2 inhibited proliferation and enhanced differentiation was also associated with a 70% increase in cell death. Although IGF-I alone was potently anti-apoptotic (60% decreased), IGF-I was unable to prevent apoptosis when administrated in combination with FGF-2. Gene-array analysis confirmed FGF-2 activation of the intrinsic and extrinsic apoptotic pathways and blockade of IGF anti-apoptotic signaling. FGF-2, directly and indirectly, overcomes the proliferative and anti-apoptotic activity of IGF-I by complex mechanisms, including enhancement of differentiation and apoptotic pathways, and inhibition of IGF-I induced anti-apoptotic signalling. Modulation of IGF binding protein abundance by FGF-2 does not play a significant role in inhibition of IGF-I induced mitogenesis.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Insulin-Like Growth Factor I/pharmacology , Neurons/drug effects , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Northern , Blotting, Western , Cell Differentiation , Cell Division/drug effects , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Gene Expression/drug effects , Humans , Immunohistochemistry , Insulin-Like Growth Factor Binding Protein 2/drug effects , Insulin-Like Growth Factor Binding Protein 6/drug effects , Neuroblastoma , Neurons/metabolism , Oligonucleotide Array Sequence Analysis
7.
Dev Dyn ; 227(3): 395-401, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12815625

ABSTRACT

The ability of glial cell line-derived neurotrophic factor (GDNF), neurturin, and artemin to induce neurite outgrowth from dorsal root, superior cervical, and lumbar sympathetic ganglia from mice at a variety of development stages between embryonic day (E) 11.5 and postnatal day (P) 7 was examined by explanting ganglia onto collagen gels and growing them in the presence of agarose beads impregnated with the different GDNF family ligands. Artemin, GDNF, and neurturin were all capable of influencing neurite outgrowth from dorsal root and sympathetic ganglia, but the responses of each neuron type to the different ligands varied during development. Neurites from dorsal root ganglia responded to artemin at P0 and P7, to GDNF at E15.5 and P0, and to neurturin at E15.5, P0, and P6/7; thus, artemin, GDNF, and neurturin are all capable of influencing neurite outgrowth from dorsal root ganglion neurons. Neurites from superior cervical sympathetic ganglia responded significantly to artemin at E15.5, to GDNF at E15.5 and P0, and to neurturin at E15.5. Neurites from lumbar sympathetic ganglia responded to artemin at all stages from E11.5 to P7, to GDNF at P0 and P7 and to neurturin at E11.5 to P6/7. Combined with the data from previous studies that have examined the expression of GDNF family members, our data suggest that artemin plays a role in inducing neurite outgrowth from young sympathetic neurons in the early stages of sympathetic axon pathfinding, whereas GDNF and neurturin are likely to be important at later stages of sympathetic neuron development in inducing axons to enter particular target tissues once they are in the vicinity or to induce branching within target tissues. Superior cervical and lumbar sympathetic ganglia showed temporal differences in their responsiveness to artemin, GDNF, and neurturin, which probably partly reflects the rostrocaudal development of sympathetic ganglia and the tissues they innervate.


Subject(s)
Ganglia, Spinal/embryology , Ganglia, Sympathetic/embryology , Gene Expression Regulation, Developmental , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurons/physiology , Animals , Axons , Cell Movement , Collagen/metabolism , Epidermal Growth Factor/metabolism , Ganglia, Sympathetic/metabolism , Glial Cell Line-Derived Neurotrophic Factor , Lumbosacral Region , Mice , Microscopy, Confocal , Nerve Growth Factors/physiology , Nerve Tissue Proteins/physiology , Neurons/metabolism , Neurturin , Time Factors
8.
Bull Math Biol ; 65(2): 235-62, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12675331

ABSTRACT

During the development of vertebrate embryos, cell migrations occur on an underlying tissue domain in response to some factor, such as nutrient. Over the time scale of days in which this cell migration occurs, the underlying tissue is itself growing. Consequently cell migration and colonization is strongly affected by the tissue domain growth. Numerical solutions for a mathematical model of chemotactic migration with no domain growth can lead to travelling waves of cells with constant velocity; the addition of domain growth can lead to travelling waves with nonconstant velocity. These observations suggest a mathematical approximation to the full system equations, allowing the method of characteristics to be applied to a simplified chemotactic migration model. The evolution of the leading front of the migrating cell wave is analysed. Linear, exponential and logistic uniform domain growths are considered. Successful colonization of a growing domain depends on the competition between cell migration velocity and the velocity and form of the domain growth, as well as the initial penetration distance of the cells. In some instances the cells will never successfully colonize the growing domain. These models provide an insight into cell migration during embryonic growth, and its dependence upon the form and timing of the domain growth.


Subject(s)
Chemotaxis/physiology , Growth/physiology , Models, Biological , Animals , Cell Count/statistics & numerical data , Cell Division/physiology , Diffusion , Numerical Analysis, Computer-Assisted , Time Factors
9.
Plast Reconstr Surg ; 108(3): 597-604, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11698829

ABSTRACT

The close spatial relationship between peripheral nerves and blood vessels in the adult is well known. However, evidence supporting the congruent development of these structures in embryos remains anecdotal. Neurovascular relationships also have been shown to be conserved in other vertebrates. This homology suggests that either peripheral nerves or blood vessels, or both, might have fundamental morphogenetic roles during embryologic development. Both peripheral nerves and blood vessels have been independently implicated as etiologic agents in the pathogenesis of congenital disabilities, and several congenital anomalies fit their distribution patterns. This article presents a technique for the simultaneous visualization of peripheral nerves and blood vessels at different stages in the developing embryo. The forelimbs of 310 quail embryos were dissected over a 1-year period. Peripheral nerves were labeled with the neural crest and axon antibody, HNK-1, followed by fluorescein-conjugated secondary antibodies. Blood vessels were labeled by a perfusion technique using the fluorescent dye, dioctadecyl-tetramethylindocarbocyanine. Specimens were processed and imaged in whole-mount with confocal microscopy, and images were reconstructed using three-dimensional modeling software. Both nerves and blood vessels seem to undergo a highly stereotypic sequence of development in the embryonic quail forelimb. Furthermore, the existence of a close spatial relationship between nerves and blood vessels suggests either a high degree of developmental interdependence or shared patterning mechanisms. This technique permits further evaluation of the possible role peripheral nerves and blood vessels might play in the pathogenesis of congenital disabilities and provides a starting point for further studies aimed at elucidating the means by which peripheral nerves and blood vessels are patterned in the forelimb of the avian embryo.


Subject(s)
Blood Vessels/embryology , Fluorescent Dyes , Microscopy, Confocal/methods , Peripheral Nerves/embryology , Animals , Histological Techniques , Immunohistochemistry , Quail
10.
Dev Biol ; 229(2): 503-16, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11150245

ABSTRACT

In situ hybridization revealed that GDNF mRNA in the mid- and hindgut mesenchyme of embryonic mice was minimal at E10.5 but was rapidly elevated at all gut regions after E11, but with a slight delay (0.5 days) in the hindgut. GDNF mRNA expression was minimal in the mesentery and in the pharyngeal and pelvic mesenchyme adjacent to the gut. To examine the effect of GDNF on enteric neural crest-derived cells, segments of E11.5 mouse hindgut containing crest-derived cells only at the rostral ends were attached to filter paper supports and grown in catenary organ culture. With GDNF (100 ng/ml) in the culture medium, threefold fewer neurons developed in the gut explants and fivefold more neurons were present on the filter paper outside the gut explants, compared to controls. Thus, in controls, crest-derived cells colonized the entire explant and differentiated into neurons, whereas in the presence of exogenous GDNF, most crest-derived cells migrated out of the gut explant. This is consistent with GDNF acting as a chemoattractant. To test this idea, explants of esophagus, midgut, superior cervical ganglia, paravertebral sympathetic chain ganglia, or dorsal root ganglia from E11.5-E12.5 mice were grown on collagen gels with a GDNF-impregnated agarose bead on one side and a control bead on the opposite side. Migrating neural cells and neurites from the esophagus and midgut accumulated around the GDNF-impregnated beads, but neural cells in other tissues showed little or no chemotactic response to GDNF, although all showed GDNF-receptor (Ret and GFRalpha1) immunoreactivity. We conclude that GDNF may promote the migration of crest cells throughout the gastrointestinal tract, prevent them from straying out of the gut (into the mesentery and pharyngeal and pelvic tissues), and promote directed axon outgrowth.


Subject(s)
Enteric Nervous System/embryology , Gene Expression Regulation, Developmental , Intestines/embryology , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Neurons/physiology , Animals , Cecum/embryology , Cecum/innervation , Cell Division/drug effects , Embryonic and Fetal Development , Glial Cell Line-Derived Neurotrophic Factor , Intestines/innervation , Mice , Nerve Growth Factors/pharmacology , Nerve Growth Factors/physiology , Nerve Tissue Proteins/physiology , Neurons/cytology , Neurons/drug effects , Organ Culture Techniques , RNA, Messenger/genetics , Transcription, Genetic
13.
Nat Genet ; 25(4): 414-8, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10932185

ABSTRACT

Secondary palate formation is a complex process that is frequently disturbed in mammals, resulting in the birth defect cleft palate. Gene targeting has identified components of cytokine/growth factor signalling systems such as Tgf-alpha/Egfr, Eph receptors B2 and B3 (Ephb2 and Ephb3, respectively), Tgf-beta2, Tgf-beta3 and activin-betaA (ref. 3) as regulators of secondary palate development. Here we demonstrate that the mouse orphan receptor 'related to tyrosine kinases' (Ryk) is essential for normal development and morphogenesis of craniofacial structures including the secondary palate. Ryk belongs to a subclass of catalytically inactive, but otherwise distantly related, receptor protein tyrosine kinases (RTKs). Mice homozygous for a null allele of Ryk have a distinctive craniofacial appearance, shortened limbs and postnatal mortality due to feeding and respiratory complications associated with a complete cleft of the secondary palate. Consistent with cleft palate phenocopy in Ephb2/Ephb3-deficient mice and the role of a Drosophila melanogaster Ryk orthologue, Derailed, in the transduction of repulsive axon pathfinding cues, our biochemical data implicate Ryk in signalling mediated by Eph receptors and the cell-junction-associated Af-6 (also known as Afadin). Our findings highlight the importance of signal crosstalk between members of different RTK subfamilies.


Subject(s)
Craniofacial Abnormalities/genetics , Receptor Cross-Talk/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Animals, Newborn , Craniofacial Abnormalities/embryology , Embryonic and Fetal Development/genetics , Female , Genotype , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mutation , Phenotype , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphB2 , Signal Transduction
14.
Hum Mol Genet ; 8(12): 2239-45, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10545604

ABSTRACT

Treacher Collins syndrome (TCS) is an autosomal dominant craniofacial disorder involving the mid and lower face and, in particular, the tissues affected arise solely from embryonic branchial arches I and II. TCOF1, the gene involved in TCS, has been cloned and although the function of the encoded protein, treacle, has not yet been established, it exhibits peak expression in the branchial arches. Treacle contains a series of repeating units of acidic and basic residues, which are predicted to contain putative casein kinase II (CKII) and protein kinase C (PKC) phosphorylation site motifs. In addition, treacle has weak homology to two phosphorylation-dependent nucleolar proteins, which shuttle between the cytoplasm and nucleolus. Based on these observations, phosphorylation of treacle may be important for its function. In this study, GST-treacle fusion peptides were constructed using particular TCOF1 exons that contained potential CKII and PKC phosphorylation sites. These were used as substrates in in vitro kinase assays and showed that treacle fusion peptides can be phosphorylated by the appropriate kinases. Furthermore, using tissue extracts we have demonstrated that in avian embryonic branchial arches I and II there is a kinase activity that can phosphorylate treacle peptides that is consistent with CKII site recognition. This activity coincides with the reported high expression of treacle in these tissues at early developmental stages and declines later in development.


Subject(s)
Branchial Region/enzymology , Mandibulofacial Dysostosis/genetics , Nuclear Proteins/genetics , Phosphoproteins/genetics , Phosphotransferases/metabolism , Animals , Base Sequence , Coturnix , DNA Primers , Glutathione Transferase/metabolism , Humans , Mandibulofacial Dysostosis/enzymology , Phosphorylation , Recombinant Fusion Proteins/genetics
15.
Cell Tissue Res ; 295(2): 195-206, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9931365

ABSTRACT

Epithelio-mesenchymal transition, which involves the re-organisation of cell-cell adhesion molecules and the actin cytoskeleton, can be induced in embryonic neural epithelium in vitro by protein kinase-C inhibitors. A non-inhibitory analogue, BIM V, and potent inhibitors of other kinases are not active. This suggests a central role for C-kinases, although the powerful specific C-kinase inhibitors BIM I and Ro 31-8220 show lower than expected activity. Co-inhibition by several kinases is unlikely to account for this, since no potentiation occurs when these are combined with potent inhibitors of other kinases. BIM I and Ro 31-8220 strongly inhibit only conventional calcium-regulated C-kinases; this and the lack of effect of TMB-8, which inhibits calcium release, suggests that novel and/or atypical isoforms are involved. Various potentiators and activators of conventional and novel C-kinases have no obvious effect alone and fail to reduce the effect of staurosporine, suggesting that atypical C-kinases are critical. The presence of C-kinase isoforms in the E2 embryonic neural tissues has been probed on Western blots, revealing immunoreactivity for the atypical isoforms iota (or lambda) and zeta and the alpha, gamma, epsilon and mu isoforms. Immunofluorecent localisation on sections of embryos has shown the widespread distribution of conventional and novel isoforms but only the atypical isoforms lambda and zeta are enriched at the apical margins of the neural and other epithelia; they overlap with the cell-cell adhesion molecule N-cadherin and with F-actin. Thus, epithelio-mesenchymal transition in the embryonic neural epithelium in vitro is induced by inhibiting protein kinase activity, probably via an atypical protein kinase-C; atypical protein kinase-C isoforms are present in the tissue at the appropriate developmental stage and subcellular site in cells capable of epithelio-mesenchymal transition.


Subject(s)
Cell Movement/physiology , Embryo, Nonmammalian/cytology , Epithelial Cells/cytology , Mesoderm/cytology , Neural Crest/cytology , Protein Kinase C/antagonists & inhibitors , Animals , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Movement/drug effects , Embryo, Nonmammalian/physiology , Enzyme Inhibitors/pharmacology , Epithelial Cells/physiology , Indoles/pharmacology , Mesoderm/physiology , Neural Crest/physiology , Quail/embryology
16.
Dev Biol ; 202(1): 67-84, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9758704

ABSTRACT

The colonization of the rodent gastrointestinal tract by enteric neuron precursors is controversial due to the lack of specific cellular markers at early stages. The transcription factor, Phox2b, is expressed by enteric neuron precursors (Pattyn et al. Development 124, 4065-4075, 1997). In this study, we have used an antiserum to Phox2b to characterize in detail the spatiotemporal expression of Phox2b in the gastrointestinal tract of adult mice and embryonic mice and rats. In adult mice, all enteric neurons (labeled with neuron-specific enolase antibodies), and a subpopulation of glial cells (labeled with GFAP antibodies), showed immunoreactivity to Phox2b. In embryonic mice, the appearance of Phox2b-immunoreactive cells was mapped during development of the gastrointestinal tract. At Embryonic Days 9.5-10 (E9.5-10), Phox2b-labeled cells were present only in the stomach, and during subsequent development, labeled cells appeared as a single rostrocaudal wave along the gastrointestinal tract; at E14 Phox2b-labeled cells were present along the entire length of the gastrointestinal tract. Ret and p75 have also been reported to label migratory-stage enteric neuron precursors. A unidirectional, rostral-to-caudal colonization of the gastrointestinal tract of embryonic mice by Ret- and p75-immunoreactive cells was also observed, and the locations of Ret- and p75-positive cells within the gut were very similar to that of Phox2b-positive cells. To verify the location of enteric neuron precursors within the gut, explants from spatiotemporally defined regions of embryonic intestine, 0.3-3 mm long, were grown in the kidney subcapsular space, or in catenary organ culture, and examined for the presence of neurons. The location and sequence of appearance of enteric neuron precursors deduced from the explants grown under the kidney capsule or in organ culture was very similar to that seen with the Phox2b, Ret, and p75 antisera. Previous studies have mapped the rostrocaudal colonization of the rat intestine by enteric neuron precursors using HNK-1 as a marker. In the current study, all HNK-1-labeled cells in the gastrointestinal tract of rat embryos showed immunoreactivity to Phox2b, but HNK-1 cells comprised only a small subpopulation of the Phox2b-labeled cells. In addition, in rats, Phox2b-labeled cells were present in advance of (more caudal to) the most caudal HNK-1-labeled cells by 600-700 microm in the hindgut at E15. We conclude that the neural crest cell population that arises from the vagal level of the neural axis and that populates the stomach, midgut, and hindgut expresses Phox2b, Ret, and p75. In contrast, the sacral-level neural crest cells that populate the hindgut either do not express, or show a delayed expression of, all of the known markers of vagal- and trunk-level neural crest cells.


Subject(s)
Drosophila Proteins , Enteric Nervous System/embryology , Homeodomain Proteins/analysis , Nerve Tissue Proteins/analysis , Neurons/chemistry , Transcription Factors/analysis , Animals , Coculture Techniques , Digestive System/embryology , Digestive System/innervation , Enteric Nervous System/cytology , Kidney Cortex/cytology , Mesoderm , Mice , Mice, Inbred BALB C , Neurons/cytology , Organ Culture Techniques , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-ret , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/analysis , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/analysis
17.
Hum Mol Genet ; 6(7): 1037-42, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9215672

ABSTRACT

The brindled mouse mutant (Mo(br)) is the closest animal model of the human genetic copper deficiency, Menkes disease, which is presumed to be due to a mutation at the X-linked mottled locus (Mo). The mutant mice are hypopigmented and die at around 15 days after birth, but can be saved by treatment with copper before the 10th postnatal day. Menkes disease has been shown to be due to mutations of the gene ATP7A which encodes P-type ATPase (referred to here as MNK). MNK is likely to function in copper efflux from cells, but the full range of its biological activity is not fully understood. The nature of the mutation in the brindled mouse is of importance in our understanding of the role of MNK and for devising treatment strategies for Menkes disease. Here we show that the brindled mouse has a deletion of two amino acids in a highly conserved, but functionally uncharacterized, region of Mnk. Comparison with the Ca ATPases suggests this region may be involved in conformational changes associated with the E1/E2 transition fundamental to the action of P-type ATPases. We also describe the first Western blot data for Mnk in tissues, and these show normal levels of Mnk in mutant and brindled kidneys but none in liver. In the kidney, immunohistochemistry demonstrated Mnk in the proximal and distal tubules, the distribution is identical in mutant and normal. This distribution is consistent with Mnk being involved in copper resorption from the urine.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cation Transport Proteins , Kidney/metabolism , Menkes Kinky Hair Syndrome/genetics , Recombinant Fusion Proteins , Sequence Deletion , Amino Acid Sequence , Animals , Blotting, Western , Conserved Sequence , Copper-Transporting ATPases , Humans , Kidney Tubules, Distal/metabolism , Kidney Tubules, Proximal/metabolism , Mice , Mice, Mutant Strains , Molecular Sequence Data , Mutation
18.
Teratology ; 55(3): 195-207, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9181673

ABSTRACT

Quail embryos (embryonic days 2-2.5) with spontaneous neural tube defects (NTDs), along with age-matched normal embryos, were examined immunocytochemically for the extracellular matrix (ECM) molecules laminin, fibronectin, and chondroitin sulfate proteoglycan, the cell adhesion molecules (CAMs) E- and N-cadherin and neural CAM (NCAM), and the neural crest marker HNK-1. The embryos with NTDs were at the lower limit of the normal stage range and the affected region was about 25% shorter than in normal embryos. Open NTDs occurred in cervical and upper thoracic level, although often the ventral neural tube was morphologically normal. Widened, irregular but closed neural tubes (lower thoracic to sacral levels) showed disorganized mesenchyme-like cells centrally and often multiple lumens. Finger-like tabs projecting from the ectoderm over the neural tube also occurred at lower thoracic to sacral levels. In open NTDs, the E-cadherin-labeled epidermis was incomplete dorsally, and was continuous with the N-cadherin-labeled neural tissue, with a sharp demarcation between E- and N-cadherin-expressing regions, as in the early stages of normal primary neurulation. A sharp inverted peak of epidermis extended ventrally, closely applied to the side of the neural tissue. The intervening matrix labeled less intensely for chondroitin sulfate proteoglycan relative to laminin and fibronectin, in comparison to control embryos. In closed NTDs, the dorsal superficial cell layer (i.e., positionally epidermis) was not separated from the underlying neural tissue by a band of matrix as in control embryos. In addition, this layer expressed E-cadherin (as in normal embryos), but coexpressed N-cadherin and NCAM, which are not normally found here at this stage. This overlap region resembled the mid-dorsal tissue at earlier stages in normal secondary neurulation in the tail-bud. The tabs of tissue appeared to be localized hypertrophy of the epidermal and neural ectoderm, and also showed codistribution of E- and N-cadherin. In all these defects, matrix molecules occurred within (rather than around) the neural and epidermal epithelia. HNK-1-labeled neural crest cells were frequently absent in regions of NTDs, in contrast to control embryos. These results show that matrix and cell adhesion molecules are disturbed in spontaneous NTDs at the time of neurulation, and therefore could be involved in the generation of the defects by altering cell adhesion-dependent morphogenetic events.


Subject(s)
Cell Adhesion Molecules/metabolism , Extracellular Matrix Proteins/metabolism , Neural Tube Defects/metabolism , Animals , Cadherins/metabolism , Chondroitin Sulfates/metabolism , Coturnix , Epidermis/metabolism , Fibronectins/metabolism , Fluorescent Antibody Technique, Indirect , Laminin/metabolism , Microscopy, Electron, Scanning , Neural Cell Adhesion Molecules/metabolism , Neural Crest/metabolism , Neural Tube Defects/pathology , Time Factors
19.
Dev Biol ; 192(1): 108-24, 1997 Dec 01.
Article in English | MEDLINE | ID: mdl-9405101

ABSTRACT

The movement of neural crest cells is controlled in part by extracellular matrix. Aggrecan, the chondroitin sulfate proteoglycan from adult cartilage, curtails the ability of neural crest cells to adhere, spread, and move across otherwise favorable matrix substrates in vitro. Our aim was to isolate, characterize, and compare the structure and effect on neural crest cells of aggrecan and proteoglycans purified from the tissues through which neural crest cells migrate. We metabolically radiolabeled proteoglycans in E2.5 quail embryos and isolated and characterized proteoglycans from E3.3 quail trunk and limb bud. The major labeled proteoglycan was highly negatively charged, similar in hydrodynamic size to chick limb bud versican/PG-M, smaller than adult cartilage aggrecan but larger than reported for embryonic sternal cartilage aggrecan. The molecular weight of the iodinated core protein was about 400 kDa, which is more than reported for aggrecan but less than that of chick versican/PG-M. The proteoglycan bore chondroitin sulfate glycosaminoglycan chains of 45 kDa, which is larger than those of aggrecan. It lacked dermatan sulfate, heparan sulfate, or keratan sulfate chains. It bound to collagen type I, like aggrecan, but not to fibronectin (unlike versican/PG-M), collagen type IV, or laminin-1 in solid-phase assays and it bound to hyaluronate in gel-shift assays. When added at concentrations between 10 and 30 microg/ml to substrates of fibronectin, trunk proteoglycan inhibited neural crest cell spreading and migration. Attenuation of cell spreading was shown to be the most sensitive and titratable measure of the effect on neural crest cells. This effect was sensitive to digestion with chondroitinase ABC. Similar cell behavior was also produced by aggrecan and the small dermatan sulfate proteoglycan decorin; however, 30-fold more aggrecan was required to produce an effect of similar magnitude. When added in solution to neural crest cells which were already spread and migrating on fibronectin, the embryonic proteoglycan rapidly and reversibly caused complete rounding of the cells, being at least 30-fold more potent than aggrecan in this activity.


Subject(s)
Chondroitin Sulfate Proteoglycans/isolation & purification , Extracellular Matrix Proteins , Neural Crest/chemistry , Neural Crest/cytology , Aggrecans , Animals , Autoradiography , Cell Adhesion/drug effects , Cell Movement/drug effects , Chondroitin Sulfate Proteoglycans/pharmacology , Chondroitin Sulfate Proteoglycans/physiology , Chromatography, Ion Exchange , Coturnix , Extracellular Matrix/metabolism , Female , Fibronectins/metabolism , In Vitro Techniques , Lectins, C-Type , Molecular Structure , Molecular Weight , Proteoglycans/isolation & purification , Proteoglycans/pharmacology , Proteoglycans/physiology , Solubility
20.
Genomics ; 38(2): 206-14, 1996 Dec 01.
Article in English | MEDLINE | ID: mdl-8954803

ABSTRACT

The neural crest is of fundamental importance in the developments of the head and peripheral nervous system, in the evolution of the vertebrates, and clinically because it gives rise to developmental abnormalities and neoplasms in humans. We have established a resource for studying the development of the neural crest by systematically constructing cDNA libraries from spatiotemporally exact neural crest and related cell populations. Neural crest populations were obtained from vagal and thoracic axial levels and from branchial arches, at premigratory and early and late migratory stages, at localization stages, and after differentiation into dorsal root ganglion cells, Schwann cells, sympathetic neurons, adrenal medullary cells, and melanocytes. Libraries were constructed using several methods developed to approach the issue of making representative libraries from small amounts of tissue. The fidelity and usefulness of the libraries were tested, and this revealed that they expressed a variety of sequences such as integrins, CAMs, growth factors and their receptors, protein-tyrosine kinases, and phosphatases. Differential display also revealed a unique combination of cDNA species. We then selected libraries spatiotemporally appropriate for epithelium-mesenchyme transformation and probed for TGF-beta-related sequences. As anticipated, we confirmed the presence of TGF-beta 2 and dorsalin-1 but could not detect TGF-beta 1. We also revealed new expression sites, defined by the origin of the libraries, of receptors known to be expressed elsewhere (Tsk 7l; TBRII). We anticipate that this collection of cDNA libraries will be of use in studying normal and abnormal neural crest development, by both homology searches and differential expression approaches, with spatiotemporal expression information being inherent in the initial screen.


Subject(s)
DNA, Complementary , Nervous System Diseases/genetics , Nervous System Neoplasms/genetics , Neural Crest/physiology , Quail/genetics , Animals , Gene Library , Neural Crest/embryology , Quail/embryology
SELECTION OF CITATIONS
SEARCH DETAIL
...