Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Mol Vis ; 14: 2575-96, 2008.
Article in English | MEDLINE | ID: mdl-19122831

ABSTRACT

PURPOSE: Asymmetric cell division (ACD) is the fundamental mechanism underlying the generation of cellular diversity in invertebrates and vertebrates. During Drosophila neuroblast division, this process involves stabilization of the apical complex and interaction between the Inscuteable (Insc) and Partner of inscuteable (Pins) proteins. Both cell-intrinsic factors and cell-cell interactions seem to contribute to cell fate decisions in the retina. The Pins protein is known to play a major role in the asymmetric segregation of cell fate determinants during development of the central nervous system in general, but its role in asymmetric cell divisions and retinoblast cell fate has never been explored. The primary aim of this study was to determine the spatial distribution and time course of mouse homolog of Drosophila Partner of Inscuteable (mPins) expression in the developing and adult mouse eye. METHODS: The expression pattern of mPins was studied in the mouse eye from embryonic (E) stage E11.5 until adulthood, by semiquantitative RT-PCR, in situ hybridization, and immunohistochemistry. In addition, variations in mRNA and protein levels for mPins were analyzed in the developing postnatal and adult lens, by semiquantitative RT-PCR, western blot analysis, in situ hybridization, and immunohistochemistry. RESULTS: We detected mPins mRNA at early stages of mouse embryonic eye development, particularly in the neuroblastic layer. In early postnatal development, mPins mRNA was still detected in the neuroblastic layer, but also began to be detectable in the ganglion cell layer. Thereafter, mPins mRNA was found throughout the retina. This pattern was maintained in differentiated adult retina. Immunohistochemical studies showed that mPins protein was present in the neuroblastic layer and the ganglion cell layer during the early stages of postnatal retinal development. At these stages, mPins protein was colocalized with Numb protein, a marker of the ACD. At later postnatal stages, mPins protein was present in all retinal nuclear layers and in the inner plexiform layer. It continued to be detected in these layers in the differentiated retina; the outer plexiform layer and the photoreceptor inner segments also began to display positive immunostaining for mPins. In the adult retina, mPins was also detected in the retinal pigment epithelium and choroidal melanocytes. Throughout development, mPins protein was detected in nonretinal tissues, including the cornea, ciliary body, and lens. We focused our attention on lens development and showed that mPins protein was first detected at E14.5. The most striking results obtained concerned the lens, in which mPins protein distribution switched from the anterior to the posterior region of the lens during embryonic development. Interestingly, in the postnatal and adult lens, mPins protein was detected in all lens cells and fibers. CONCLUSIONS: We provide the first demonstration that mPins protein is expressed from embryonic stages until adulthood in the mouse eye. These results suggest that mPins plays important roles in eye development. This work provides preliminary evidence strongly supporting a role for mPins in the asymmetric division of retinoblasts, and in the structure and functions of adult mouse retina. However, the link between the presence of mPins in different ocular compartments and the possible occurrence of asymmetric cell divisions in these compartments remains to be clarified. Further studies are required to elucidate the in vitro and in vivo functions of mPins in the developing and adult human eye.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Eye/embryology , Eye/metabolism , Gene Expression Regulation, Developmental , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Newborn , Antibodies , Cell Differentiation , Ciliary Body/cytology , Ciliary Body/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Eye/cytology , Immunohistochemistry , In Situ Hybridization , Lens, Crystalline/cytology , Lens, Crystalline/embryology , Lens, Crystalline/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Protein Transport , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/cytology , Retina/embryology , Retina/metabolism
2.
Mol Vis ; 13: 1412-27, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17768378

ABSTRACT

PURPOSE: Musashi-1 (Msi1) is an RNA-binding protein produced in various types of stem cells including neural stem/progenitor cells and astroglial progenitor cells in the vertebrate central nervous system. Other RNA-binding proteins such as Pumilio-1, Pumilio-2, Staufen-1, and Staufen-2 have been characterized as potential markers of several types of stem or progenitor cells. We investigated the involvement of Msi1 in mouse eye development and adult mouse eye functions by analyzing the profile of Msi1 production in all ocular structures during development and adulthood. METHODS: We studied Msi1 production by in situ hybridization and immunohistochemistry of ocular tissue sections and by semi-quantitative RT-PCR and western blot analysis from the embryonic stage of 12.5 days post coitum (E12.5 dpc) when the first retinal ganglion cells (RGCs) begin to appear to the adult stage when all retinal cell types are present. RESULTS: Msi1 mRNA was present at all studied stages of eye development. Msi1 protein was detected in the primitive neuroblastic layer (NbL), the ganglion cell layer (GCL), and in all major differentiated neurons of postnatal developing and adult retinae. During postnatal developing stages, faint diffuse Msi1 protein staining is converted to a more specific distribution once mouse retina is fully differentiated. The most striking result of our study concerns the large amounts of Msi1 protein and mRNA in several unexpected sites of adult mouse eyes including the corneal epithelium and endothelium, stromal keratocytes, progenitor cells of the limbus, equatorial lens stem cells, differentiated lens epithelial cells, and differentiating lens fibers. Msi1 was also found in the pigmented and nonpigmented cells of the ciliary processes, the melanocytes of the ciliary body, the retinal pigment epithelium, differentiated retinal neurons, and most probably in the retinal glial cells such as Müller glial cells, astrocytes, and the oligodendocytes surrounding the axons of the optic nerve. Msi1 expression was detected in the outer plexiform layer, the inner plexiform layer, and the nerve fiber layer of fully differentiated adult retina. CONCLUSIONS: We provide here the first demonstration that the RNA-binding protein, Msi1, is produced in mouse eyes from embryonic stages until adulthood. The relationship between the presence of Msi1 in developing ocular compartments and the possible stem/progenitor cell characteristics of these compartments remains unclear. Finally, the expression of Msi1 in several different cell types in the adult eye is extremely intriguing and should lead to further attempts to unravel the role of Msi1 in cellular and subcellular RNA metabolism and in the control of translational processes in adult eye cells particularly in adult neuronal dendrites, axons, and synapses.


Subject(s)
Eye/embryology , Eye/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Animals , Animals, Newborn , Antibody Specificity , Eye/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism
3.
Cell Commun Adhes ; 8(3): 99-112, 2001.
Article in English | MEDLINE | ID: mdl-11936189

ABSTRACT

The alpha v beta (alpha(v)beta5) heterodimer has been implicated in many biological functions, including angiogenesis. We report the beta5 gene expression pattern in embryonic and foetal mouse tissues as determined by Northern blotting and in situ hybridization. During the earliest stages, beta5 mRNA is widespread in the mesoderm. During later developmental stages, it remains mostly confined to tissues of mesodermal origin, although probable inductive effects trigger shifts of beta5 gene expression from some mesenchymatous to epithelial structures. This was observed in the teeth, skin, kidneys, and gut. Of physiological importance is the beta5 labeling in the developing cardiovascular and respiratory systems and cartilages. Furthermore, early beta5 gene expression was observed within the intra- and extraembryonic sites of hematopoiesis. This suggests a major role for beta5 in the hematopoietic and angiogenic stem cells and thus in the development of the vascular system. Later, the beta5 gene was expressed in endothelial cells of the vessels developing both by angiogenesis and vasculogenesis in the lung, heart, and kidneys. Moreover, the beta5 hybridization signal was detected in developing cartilages but not in ossified or ossifying bones. beta5-Integrin is a key integrin involved in angiogenesis, vasculogenesis, hematopoiesis, and bone formation.


Subject(s)
Bone and Bones/embryology , Cardiovascular System/embryology , Cartilage/embryology , Integrin beta Chains/metabolism , Mice/embryology , Animals , Base Sequence , Blotting, Northern , Cardiovascular System/metabolism , Cartilage/metabolism , Digestive System/metabolism , Digestive System/ultrastructure , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , In Situ Hybridization , Integrin beta Chains/genetics , Kidney/growth & development , Kidney/ultrastructure , Mice/genetics , Mice/metabolism , Molecular Sequence Data , Respiratory System/metabolism
4.
Hum Gene Ther ; 11(13): 1875-90, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10986560

ABSTRACT

Based on the K8/JTS-1-mediated transfection technique, we developed an in vivo protocol for an efficient transfer of plasmid DNA to ocular cells. As determined with condensed plasmids containing reporter genes for either beta-galactosidase (pcDNA-lacZ) or enhanced green fluorescent protein (pREP-EGFP), the immortalized human retinal epithelial cells RPE D407 and human embryonic kidney 293 cells can be transfected with typical efficiencies of 11 and 19%, respectively. Unlike 293 cells, RPE D407 cells had a reduced viability on transfection with both plasmids. In vivo, subretinal injections of DNA-K8/JTS-1 complexes revealed reporter gene expression in choroidal and RPE cells of normal pink-eyed Royal College of Surgeons (RCS) rats. The validity of this transfection technique in terms of retinal cell survival in RCS rats was then examined by using pREP-hFGF2 plasmid, which encodes the human basic fibroblast growth factor isoforms (hFGF2). Subretinal injection of pREP-hFGF2-K8/JTS-1 complexes into 3-week-old dystrophic RCS rat eyes reveals a delayed photoreceptor cell degeneration 60 days postinjection. In this case, the average analyzed field points with delayed cell dystrophy represent 14 to 17% of the retinal surface as compared with 2.6 and 4% in pREP5beta and vehicle-injected eyes, respectively. Peptide-mediated in oculo transfection thus appears to be a promising technique for the treatment of retinal cell and photoreceptor degenerations.


Subject(s)
Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/pharmacology , Genetic Therapy/methods , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/genetics , Transfection/methods , Animals , Base Sequence , Cell Survival/genetics , Fibroblast Growth Factor 2/metabolism , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microinjections , Molecular Sequence Data , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/physiology , Pigment Epithelium of Eye/cytology , Plasmids/genetics , Rats , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
5.
J Neuropathol Exp Neurol ; 58(8): 867-80, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10446811

ABSTRACT

We analyzed the distribution of FMR1, FXR1, FXR2 mRNA, and FMRP in whole normal human embryos and in the brains of normal and fragile X fetuses. The distributions of mRNA for the 3 genes in normal whole embryos and in the brains of normal male and female carrier fetuses were similar, with large amounts of mRNA in the nervous system and in several non-nervous system tissues. No FMR1 (mRNA and protein) was detected and no evident neuropathologic abnormalities found in the brains of male carrier fetuses, suggesting that the FMR1 product (FMRP) may have no crucial function in early stages of nervous system development. FXR1 and FXR2 mRNA had the same distribution and similar intensity in the brains of normal and pathologic fetuses (female and male carriers). The coexpression in the same tissues of FMR1, FXR1, and FXR2, associated with the normal expression of FXR1 and FXR2 and the absence of obvious neuropathological abnormalities in pathological brains, supports the notion that the FXR1 and FXR2 proteins partially compensate for FMRP function. However, the absence of significant overexpression of FXR1 and FXR2 in pathological brains suggests that these genes do not compensate for the lack of FMR1 expression. Alternatively, FMR1, FXR1, and FXR2 proteins may not have compensatory functions, but instead may regulate functions by hetero or homo oligomerization, as suggested by other studies. Thus, a dominant negative effect of abnormal multimeric protein complexes lacking FMRP (e.g. by modification of FXR1 and FXR2 protein functions) may result in the fragile X syndrome phenotype.


Subject(s)
Fragile X Syndrome/metabolism , Gene Expression Regulation, Developmental/physiology , Nerve Tissue Proteins/biosynthesis , RNA-Binding Proteins/biosynthesis , Base Sequence , Blotting, Northern , Blotting, Western , Female , Fragile X Mental Retardation Protein , Fragile X Syndrome/genetics , Gestational Age , Humans , Immunohistochemistry , In Situ Hybridization , Male , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Oligonucleotide Probes , Pregnancy , RNA, Messenger/biosynthesis , RNA-Binding Proteins/genetics
6.
Proc Natl Acad Sci U S A ; 96(6): 3126-31, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077648

ABSTRACT

We developed an experimental approach with genetically engineered and encapsulated mouse NIH 3T3 fibroblasts to delay the progressive degeneration of photoreceptor cells in dark-eyed Royal College of Surgeons rats. These xenogeneic fibroblasts can survive in 1. 5-mm-long microcapsules made of the biocompatible polymer AN69 for at least 90 days under in vitro and in vivo conditions because of their stable transfection with the gene for the 18-kDa form of the human basic fibroblast growth factor (hFGF-2). Furthermore, when transferred surgically into the vitreous cavity of 21-day-old Royal College of Surgeons rats, the microencapsulated hFGF-2-secreting fibroblasts provoked a local delay of photoreceptor cell degeneration, as seen at 45 days and 90 days after transplantation. This effect was limited to 2.08 mm2 (45 days) and 0.95 mm2 (90 days) of the retinal surface. In both untreated eyes and control globes with encapsulated hFGF-2-deficient fibroblasts, the rescued area (of at most 0.08 mm2) was significantly smaller at both time points. Although, in a few ocular globes, surgical trauma induced a reorganization of the retinal cytoarchitecture, neither microcapsule rejection nor hFGF-2-mediated tumor formation were detected in any treated eyes. These findings indicate that encapsulated fibroblasts secreting hFGF-2 or perhaps other agents can be applied as potential therapeutic tools to treat retinal dystrophies.


Subject(s)
Fibroblast Growth Factor 2/genetics , Fibroblasts/transplantation , Gene Transfer Techniques , Genetic Therapy , Photoreceptor Cells, Vertebrate/pathology , Retinal Degeneration/therapy , 3T3 Cells , Acrylic Resins , Acrylonitrile/analogs & derivatives , Animals , Cell Death , Fibroblast Growth Factor 2/metabolism , Fibroblasts/metabolism , Humans , Mice , Rats , Retinal Degeneration/genetics , Retinal Degeneration/pathology
8.
Brain Res Mol Brain Res ; 58(1-2): 123-31, 1998 Jul 15.
Article in English | MEDLINE | ID: mdl-9685607

ABSTRACT

Neuronal nicotinic acetylcholine receptors (nAChRs) present in the central nervous system (CNS), are multimeric proteins constituted of two different subunits, alpha and beta, with different subtype arrangements and different pharmacological and functional properties. By in situ hybridization, we studied the distribution of the mRNA for the alpha4 subunit of nAChRs in brains of human 25-week old normal and fragile X fetuses. A strong hybridization signal was detected throughout the thalamus, cortex, pyramidal layer of the Ammon's horn, and the granular layer of the dentate gyrus. Several other areas including the claustrum, caudate nucleus, putamen, globus pallidus, subthalamic nucleus, subiculum, entorhinal cortex, and Purkinje cell layer displayed a low to moderate radiosignal. With few exceptions, our data in the human brain agree those previously reported in the rat. Also, our data indicate that the alpha4 subunit mRNA is produced early in the development, in the more differentiated cells, and in a site-specific manner. Additionally, the alpha4 mRNA is produced in the brain of fragile X fetuses with the same pattern and same intensity than in the normal fetal brain suggesting that alpha4 subunit mRNA production is not altered in the fragile X syndrome. High levels of alpha4 subunit mRNA in human fetal brain support the hypothesis of a morphogenic role of nAChRs during the early CNS development.


Subject(s)
Brain/embryology , Neurons/metabolism , RNA, Messenger/analysis , Receptors, Nicotinic/biosynthesis , Animals , Autoradiography , Brain/metabolism , Brain/pathology , Female , Fetus , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Humans , In Situ Hybridization , Macromolecular Substances , Male , Oligonucleotides, Antisense , Organ Specificity , Pregnancy , Purkinje Cells/metabolism , Rats , Receptors, Nicotinic/chemistry , Sulfur Radioisotopes
9.
Hum Mol Genet ; 7(9): 1437-47, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9700199

ABSTRACT

The tubby strain of mice exhibits maturity-onset obesity and sensory deficits in vision and hearing. The mutated gene, tub , responsible for this phenotype was identified recently, but the function of the TUB protein has not been deduced from its amino acid sequence. This prompted us to undertake expression mapping studies with the hope that they might help to elucidate the biological role of the TUB protein. We report the tub gene expression pattern in embryonic, fetal and adult mice tissues as determined by northern blots and in situ hybridization, using antisense oligonucleotidic probes. In mouse embryos, tub is expressed selectively in differentiating neurons of the ensemble of central and peripheral nervous systems, starting at 9.5 days after conception. In adult mice, tub is transcribed in several major brain areas, including cerebral cortex, hippocampus, several nuclei of the hypothalamus controlling feeding behavior, in the spiral ganglion of the inner ear and in the photoreceptor cells of the retina. These structures contain potential cellular targets of the tubby mutation-induced pathogenesis. The neuronal-specific tub gene distribution allows the establishment of a genotype-phenotype correlation in the tubby mice. This correlation is reminiscent of that observed in fat/fat mice, whose phenotype, also characterized by obesity, is caused by a null mutation in the carboxypeptidase E (CPE) gene. Our observations highlight similarities between CPE, prohormone convertases, several neuropeptides and tub gene expression patterns during embryogenesis, and may narrow down the avenues to explore in order to determine ultimately the function of the TUB protein.


Subject(s)
Mutation , Proteins/genetics , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Female , Gene Expression Regulation, Developmental , Hearing Disorders/genetics , In Situ Hybridization , Mice , Neurons/metabolism , Obesity/genetics , Oligonucleotide Probes/genetics , Pregnancy , Vision Disorders/genetics
10.
FEBS Lett ; 432(3): 168-72, 1998 Aug 07.
Article in English | MEDLINE | ID: mdl-9720918

ABSTRACT

SOX10, a new member of the SOX gene family, is a transcription factor defective in the Dom (Dominant megacolon) mouse and in the human Shah-Waardenburg syndrome. To help unravel its physiological role during human development, we studied SOX10 gene expression in embryonic, fetal, and adult human tissues by Northern blot and in situ hybridization. As in mice, the human SOX10 gene was essentially expressed in the neural crest derivatives that contribute to the formation of the peripheral nervous system, and in the adult central nervous system. Nevertheless, it was more widely expressed in humans than in rodents. The spatial and temporal pattern of SOX10 expression supports an important function in neural crest development.


Subject(s)
DNA-Binding Proteins/genetics , Embryo, Mammalian/chemistry , Embryonic and Fetal Development/genetics , High Mobility Group Proteins/genetics , Blotting, Northern , Central Nervous System/chemistry , Central Nervous System/embryology , Gene Expression/genetics , Humans , In Situ Hybridization , Neural Crest/chemistry , Neural Crest/cytology , Neural Crest/embryology , RNA, Messenger/analysis , RNA, Messenger/genetics , SOXE Transcription Factors , Tissue Distribution , Transcription Factors/genetics
11.
Am J Hum Genet ; 62(5): 1117-22, 1998 May.
Article in English | MEDLINE | ID: mdl-9545411

ABSTRACT

Congenital microcoria is an autosomal dominant disorder characterized by a pupil with a diameter <2 mm. It is thought to be due to a maldevelopment of the dilator pupillae muscle of the iris, and it is associated with juvenile-onset glaucoma. A total genome search for the location of the congenital microcoria gene was launched in a single large family. We found linkage between the disease and markers located on 13q31-q32 (Zmax = 9.79; theta = 0). Haplotype analysis narrowed the linked region to an interval <8 cM between markers D13S1239 proximally and D13S1280 distally.


Subject(s)
Chromosomes, Human, Pair 13 , Iris/abnormalities , Chromosome Mapping , Female , Genetic Linkage , Haplotypes , Humans , Male , Pedigree , Recombination, Genetic
12.
Hum Mutat ; 12(2): 138, 1998.
Article in English | MEDLINE | ID: mdl-10694925

ABSTRACT

We report two novel PAX6 mutations in aniridia patients of two Swiss pedigrees (We, Sc) which give rise to different phenotypes. An SSCP analysis of the PAX6 14 exons reveals electrophoretic mobility shifts exclusively in exons 5 and 12 of aniridia patients. As determined by bidirectional sequencing and restriction digest analysis, these shifts are caused by mono-allelic base transitions in exon 5 (c.547C-->T; R44X; We) and intron 12 (IVS12+5G-->A; Sc). Each mutation co-segregates with the trait in the affected family with complete penetrance. The Sc mutation in the splicing donor site of intron 12 may result in either intron inclusion or exon skipping, both giving rise to a truncated PAX6 protein which may retain a residual transactivating activity. In contrast, the We genetic alteration is a loss-of-function mutation leading to a more severe phenotype than that observed in the Sc pedigree.


Subject(s)
Aniridia/genetics , DNA-Binding Proteins/genetics , Homeodomain Proteins , Mutation/genetics , Alternative Splicing , Eye Proteins , Humans , Introns/genetics , PAX6 Transcription Factor , Paired Box Transcription Factors , Pedigree , Repressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...