Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Ann Dermatol Venereol ; 129(5 Pt 2): 793-9, 2002 May.
Article in French | MEDLINE | ID: mdl-12223961

ABSTRACT

The active metabolite of vitamin A (retinoic acid, RA) acts through the nuclear receptors RARalpha, beta and gamma and RXRalpha, beta and gamma. These receptors form RAR/RXR heterodimers, which bind to genetic regulatory DNA sequences and activate transcription of RA target genes. As RXR form heterodimers with a number of other nuclear receptors, such as the vitamin D3 receptor (VDR) and are involved in several signaling pathways. In the skin, RARgamma and RXRalpha predominate, but RARalpha and RXRbeta are also expressed. To elucidate the role of RA in skin physiology, we produced mutant mouse lines null for RAR or RXR. On the one hand, null mutations for RARa or RXRbeta have no effect on the skin, whereas a RARgamma-null mutation induces alterations in the granular cell layer. On the other, genetic inactivation of RXRa leads to embryonic lethality before epidermal development. Consequently, to determine the role of RXRa in adult mice, studies were performed using conditional somatic mutagenesis (permitting inactivation of a given gene in a specific tissue and in a time-dependent manner). Using this novel genetic approach, mutant mice were obtained in which RXRalpha was not expressed in the skin. These mice developed hair follicle degeneration, then alopecia, similar to that observed in VDR-null mutants, suggesting that hair follicle homeostasis depends on RXRalpha/VDR heterodimers. A similar genetic approach applied to the RARgamma locus demonstrated that topical administration of RA on the skin activates RARgamma/RXR heterodimers in suprabasal cells, and induces expression of a paracrine growth factor (HB-EGF) in these cells which, in turn, stimulates the proliferation of basal cells.


Subject(s)
Keratolytic Agents/pharmacology , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/physiology , Skin Physiological Phenomena , Tretinoin/pharmacology , Alopecia/genetics , Alopecia/physiopathology , Animals , DNA/biosynthesis , Gene Expression Regulation , Hair Follicle/physiology , Mice , Point Mutation , Signal Transduction
2.
Development ; 128(11): 2031-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11493525

ABSTRACT

Mutants mice carrying targeted inactivations of both retinoic acid receptor (RAR) alpha and RAR gamma (A alpha/A gamma mutants) were analyzed at different embryonic stages, in order to establish the timing of appearance of defects that we previously observed during the fetal period. We show that embryonic day (E)9.5 A alpha/A gamma embryos display severe malformations, similar to those already described in retinaldehyde dehydrogenase 2 null mutants. These malformations reflect early roles of retinoic acid signaling in axial rotation, segmentation and closure of the hindbrain; formation of otocysts, pharyngeal arches and forelimb buds; and in the closure of the primitive gut. The hindbrain of E8.5 A alpha/A gamma embryos shows a posterior expansion of rhombomere 3 and 4 (R3 and R4) markers, but fails to express kreisler, a normal marker of R5 and R6. This abnormal hindbrain phenotype is strikingly different from that of embryos lacking RAR alpha and RAR beta (A alpha/A beta mutants), in which we have previously shown that the territory corresponding to R5 and R6 is markedly enlarged. Administration of a pan-RAR antagonist at E8.0 to wild-type embryos cultured in vitro results in an A alpha/A beta-like hindbrain phenotype, whereas an earlier treatment at E7.0 yields an A alpha/A gamma-like phenotype. Altogether, our data suggest that RAR alpha and/or RAR gamma transduce the RA signal that is required first to specify the prospective R5/R6 territory, whereas RAR beta is subsequently involved in setting up the caudal boundary of this territory.


Subject(s)
Avian Proteins , Body Patterning , Oncogene Proteins , Receptors, Retinoic Acid/physiology , Rhombencephalon/embryology , Animals , DNA-Binding Proteins/genetics , Early Growth Response Protein 2 , Embryonic and Fetal Development , Homeodomain Proteins/genetics , MafB Transcription Factor , Mice , Morphogenesis , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Transcription Factors/genetics , Retinoic Acid Receptor gamma
3.
Development ; 128(11): 2049-62, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11493527

ABSTRACT

We have engineered a mouse mutation that specifically deletes most of the RXR alpha N-terminal A/B region, which includes the activation function AF-1 and several phosphorylation sites. The homozygous mutants (RXR alpha af1(o)), as well as compound mutants that further lack RXR beta and RXR gamma, are viable and display a subset of the abnormalities previously described in RXR alpha-null mutants. In contrast, RXR alpha af1(o)/RAR(-/-)(alpha, beta or gamma) compound mutants die in utero and exhibit a large array of malformations that nearly recapitulate the full spectrum of the defects that characterize the fetal vitamin A-deficiency (VAD) syndrome. Altogether, these observations indicate that the RXR alpha AF-1 region A/B is functionally important, although less so than the ligand-dependent activation function AF-2, for efficiently transducing the retinoid signal through RAR/RXR alpha heterodimers during embryonic development. Moreover, it has a unique role in retinoic acid-dependent involution of the interdigital mesenchyme. During early placentogenesis, both the AF-1 and AF-2 activities of RXR alpha, beta and gamma appear to be dispensable, suggesting that RXRs act as silent heterodimeric partners in this process. However, AF-2 of RXR alpha, but not AF-1, is required for differentiation of labyrinthine trophoblast cells, a late step in the formation of the placental barrier.


Subject(s)
Receptors, Retinoic Acid/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Amino Acid Sequence , Animals , Base Sequence , Gene Targeting , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutagenesis , Promoter Regions, Genetic , Receptors, Retinoic Acid/genetics , Retinoid X Receptors , Trans-Activators/genetics , Transcription Factors/genetics
4.
Invest Ophthalmol Vis Sci ; 42(6): 1312-8, 2001 May.
Article in English | MEDLINE | ID: mdl-11328745

ABSTRACT

PURPOSE: To determine the localization of retinoic acid receptors (RAR) alpha, beta, and gamma and retinoid X receptors (RXR) alpha, beta, and gamma in developing and adult mouse eyes at the level of single cells. METHODS: Immunohistochemistry was performed on paraformaldehyde-lysine-periodate-fixed cryosections of mouse eyes, from embryonic day 10.5 to adulthood, with polyclonal antibodies directed against each receptor isoform. Histologic sections from null mutant mice for each receptor served as negative controls. RESULTS: RARalpha was present ubiquitously in the prenatal eye and preferentially located in the posnatal retina and ciliary body. RARbeta was detected predominantly in the periocular mesenchyme and ciliary body. RARgamma was distributed in the periocular mesenchyme, choroid, sclera, cornea, conjunctiva, and lids. RXRalpha was found preferentially in the prenatal periocular mesenchyme and retina and in the postnatal ciliary body, cornea, and conjunctiva. RXRbeta was ubiquitous at all the stages. RXRgamma was detected mainly in subsets of prenatal retinal cells and in postnatal ganglion cells as well as a subset of photoreceptor cells that were characterized as cones in adults. CONCLUSIONS: RARalpha, beta, and gamma and RXRalpha and gamma exhibit specific and dynamic patterns of distribution in ocular tissues throughout the course of development. The abundance of RARbeta, RARgamma, and RXRalpha in the periocular mesenchyme suggests that this tissue represents an important site of retinoid actions during eye development and in adulthood.


Subject(s)
Eye/embryology , Receptors, Retinoic Acid/analysis , Transcription Factors/analysis , Animals , Conjunctiva/chemistry , Conjunctiva/embryology , Cornea/chemistry , Cornea/embryology , Embryonic and Fetal Development , Eye/chemistry , Eyelids/chemistry , Eyelids/embryology , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Lens, Crystalline/chemistry , Lens, Crystalline/embryology , Mice , Pigment Epithelium of Eye/chemistry , Pigment Epithelium of Eye/embryology , Retinal Cone Photoreceptor Cells/chemistry , Retinal Cone Photoreceptor Cells/embryology , Retinoic Acid Receptor alpha , Retinoid X Receptors , Retinoic Acid Receptor gamma
5.
Blood ; 97(5): 1314-20, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222375

ABSTRACT

Acute promyelocytic leukemia (APL) is always associated with chromosomal translocations that disrupt the retinoic acid receptor alpha (RARalpha) gene. Whether these translocations relate to a role for endogenous RARalpha in normal granulopoiesis remains uncertain because most studies addressing this question have used non-physiological overexpression systems. Granulocyte differentiation in cells derived from RARalpha-deficient (RARalpha(-/-)) mice was studied and evaluated in the context of agonist-bound and ligand-free RARalpha. Our results demonstrate that RARalpha is dispensable for granulopoiesis, as RARalpha(-/-) mice have a normal granulocyte population despite an impaired ability to respond to retinoids. However, although it is not absolutely required, RARalpha can bidirectionally modulate granulopoiesis. RARalpha stimulates differentiation in response to exogenous retinoic acid. Furthermore, endogenous retinoids control granulopoiesis in vivo, as either vitamin A-deficient mice or animals treated with an RAR antagonist accumulate more immature granulocytes in their bone marrow. Conversely, RARalpha acts to limit differentiation in the absence of ligand because granulocyte precursors from RARalpha(-/-) mice differentiate earlier in culture. Thus, the block in granulopoiesis exerted by RARalpha fusion proteins expressed in APL cells may correspond to an amplification of a normal function of unliganded RARalpha.


Subject(s)
Granulocytes/cytology , Granulocytes/drug effects , Receptors, Retinoic Acid/physiology , Animals , Bone Marrow Cells , Cell Culture Techniques , Cell Differentiation/drug effects , Mice , Mice, Mutant Strains , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Retinoids/pharmacology , Retinol-Binding Proteins/genetics , Retinol-Binding Proteins/pharmacology , Retinol-Binding Proteins, Cellular , Tretinoin/pharmacology , Vitamin A Deficiency
6.
Int J Dev Biol ; 44(5): 457-62, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11032179

ABSTRACT

Morphological analysis of vitamin A-deficient rat fetuses and of retinoic acid receptor (RAR and RXR) mutant mice have demonstrated that retinoic acid (RA) is essential for lung development. To gainfurther insight into RA signaling pathways during primary lung budformation and lung branching, we have investigated the effects of RA and of a pan-RAR antagonist in cultures of whole embryos and lung explants. Treatment of E8.0 embryos with the pan-RAR antagonist inhibits the formation of the primitive respiratory system. On the other hand, treatment of E11.75 and E12.5 lung explants with RA inhibits branching morphogenesis, whereas treatment with the pan-RAR antagonist at the same developmental stages stimulates formation of distal buds. The inhibitory effect of RA on branching is strongly decreased in RARbeta null lungs, while enhancement of budding by the pan-RAR antagonist is not affected by an RARgamma null mutation. Additionally, cellular retinol binding protein one (CRBPI) null lungs are more sensitive than wild type lungs to the pan-RAR antagonist-induced stimulation of branching. These data indicate that retinoid signaling is indispensable for the formation of primary lung buds and the oesophagotracheal septum from the primitive foregut. They also suggest that at the pseudoglandular stage, RA signaling through RARbeta, but not RARgamma, inhibits distal bud formation thereby promoting the formation of conducting airways. Moreover, the level of CRBPI in the pseudoglandular lung appears to participate in the control of branching morphogenesis.


Subject(s)
Lung/drug effects , Lung/embryology , Signal Transduction , Tretinoin/pharmacology , Animals , Dose-Response Relationship, Drug , In Situ Hybridization , Mutation , Organ Culture Techniques , Protein Isoforms , Rats , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Retinoic Acid/genetics , Retinol-Binding Proteins/biosynthesis , Retinol-Binding Proteins/physiology , Retinol-Binding Proteins, Cellular , Ribonucleases/metabolism , Time Factors , Retinoic Acid Receptor gamma
7.
Eur J Neurosci ; 12(8): 2793-804, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10971621

ABSTRACT

The expression patterns of the mouse cellular retinoid binding protein genes were investigated by in situ hybridization analysis in the inner ear from 10.5 days post coïtum (dpc) up to the adult stage. The cellular retinoic acid binding protein II (CRABPII) and cellular retinol binding protein I (CRBPI) were present in a widespread and abundant pattern in cochlear structures during embryogenesis. Expression of the cellular retinoic acid binding protein I (CRABPI) is restricted during development in Kölliker's organ whilst cellular retinol binding protein II (CRBPII) is only visible after birth with a ubiquitous distribution in most regions of the cochlea including nervous components. No CRABP or CRBP transcripts were observed in the auditory receptors. Morphological observations of CRBPI- and CRABPI/CRABPII-null mutant fetus at 18.5 dpc do not show any structural modification at the level of the organ of Corti. Furthermore, electrophysiological tests performed by measuring distorsion-product otoacoustic emissions and auditory brainstem evoked responses did not present significant alteration of the auditory function for the different types of mutants. The expression of retinoid binding proteins in cochlear structures during embryogenesis could suggest important roles for these proteins during ontogenesis and morphogenesis of the inner ear. Despite these observations, morphological and functional data from mutant mice did not present obvious modifications of the cochlear structures and auditory thresholds. It is therefore unlikely that CRABPs and CRBPI are directly involved in development of the cochlea and hair cell differentiation.


Subject(s)
Cochlea/growth & development , Cochlea/physiology , Receptors, Retinoic Acid/genetics , Retinol-Binding Proteins/genetics , Age Factors , Animals , Audiometry, Pure-Tone , Auditory Threshold/physiology , Cochlea/cytology , Evoked Potentials, Auditory, Brain Stem/physiology , Gene Expression Regulation, Developmental , Hair Cells, Auditory, Inner/chemistry , Hair Cells, Auditory, Inner/physiology , Hair Cells, Auditory, Outer/chemistry , Hair Cells, Auditory, Outer/physiology , In Situ Hybridization , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , RNA, Messenger/analysis , Retinol-Binding Proteins, Cellular , Transcription, Genetic/physiology
8.
Proc Nutr Soc ; 58(3): 609-13, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10604193

ABSTRACT

To determine the functions of retinoic acid receptors RAR and RXR, we have systematically knocked-out their genes by homologous recombination in the embryonic stem cells and generated null-mutant mice. This approach has allowed us to perform a genetic dissection of the retinoic acid signalling pathway.


Subject(s)
Retinoids , Signal Transduction/genetics , Animals , Mice , Mice, Knockout , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/physiology , Retinoid X Receptors , Transcription Factors/genetics , Transcription Factors/physiology
9.
Development ; 126(22): 5051-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10529422

ABSTRACT

Mouse fetuses carrying targeted inactivations of both the RAR(&agr;) and the RARbeta genes display a variety of malformations in structures known to be partially derived from the mesenchymal neural crest originating from post-otic rhombomeres (e.g. thymus and great cephalic arteries) (Ghyselinck, N., Dupé, V., Dierich, A., Messaddeq, N., Garnier, J.M., Rochette-Egly, C., Chambon, P. and Mark M. (1997). Int. J. Dev. Biol. 41, 425-447). In a search for neural crest defects, we have analysed the rhombomeres, cranial nerves and pharyngeal arches of these double null mutants at early embryonic stages. The mutant post-otic cranial nerves are disorganized, indicating that RARs are involved in the patterning of structures derived from neurogenic neural crest, even though the lack of RARalpha and RARbeta has no detectable effect on the number and migration path of neural crest cells. Interestingly, the double null mutation impairs early developmental processes known to be independent of the neural crest e.g., the initial formation of the 3rd and 4th branchial pouches and of the 3rd, 4th and 6th arch arteries. The double mutation also results in an enlargement of rhombomere 5, which is likely to be responsible for the induction of supernumerary otic vesicles, in a disappearance of the rhombomere 5/6 boundary, and in profound alterations of rhombomere identities. In the mutant hindbrain, the expression domain of kreisler is twice its normal size and the caudal stripe of Krox-20 extends into the presumptive rhombomeres 6 and 7 region. In this region, Hoxb-1 is ectopically expressed, Hoxb-3 is ectopically up-regulated and Hoxd-4 expression is abolished. These data, which indicate that retinoic acid signaling through RARalpha and/or RARbeta is essential for the specification of rhombomere identities and for the control of caudal hindbrain segmentation by restricting the expression domains of kreisler and of Krox-20, also strongly suggest that this signaling plays a crucial role in the posteriorization of the hindbrain neurectoderm.


Subject(s)
Avian Proteins , Branchial Region/embryology , Oncogene Proteins , Receptors, Retinoic Acid/physiology , Rhombencephalon/embryology , Animals , Branchial Region/abnormalities , Cell Count , Cell Movement , Cleavage Stage, Ovum/physiology , Cranial Nerves/abnormalities , Cranial Nerves/embryology , DNA-Binding Proteins/biosynthesis , Early Growth Response Protein 2 , Embryonic and Fetal Development , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , MafB Transcription Factor , Mice , Neural Crest/cytology , Receptors, Retinoic Acid/genetics , Retinoic Acid Receptor alpha , Rhombencephalon/abnormalities , Signal Transduction , Transcription Factors/biosynthesis , Tretinoin/metabolism
10.
EMBO J ; 18(18): 4903-14, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10487743

ABSTRACT

The gene encoding cellular retinol (ROL, vitA)-binding protein type I (CRBPI) has been inactivated. Mutant mice fed a vitA-enriched diet are healthy and fertile. They do not present any of the congenital abnormalities related to retinoic acid (RA) deficiency, indicating that CRBPI is not indispensable for RA synthesis. However, CRBPI deficiency results in an approximately 50% reduction of retinyl ester (RE) accumulation in hepatic stellate cells. This reduction is due to a decreased synthesis and a 6-fold faster turnover, which are not related to changes in the levels of RE metabolizing enzymes, but probably reflect an impaired delivery of ROL to lecithin:retinol acyltransferase. CRBPI-null mice fed a vitA-deficient diet for 5 months fully exhaust their RE stores. Thus, CRBPI is indispensable for efficient RE synthesis and storage, and its absence results in a waste of ROL that is asymptomatic in vitA-sufficient animals, but leads to a severe syndrome of vitA deficiency in animals fed a vitA-deficient diet.


Subject(s)
Retinol-Binding Proteins/genetics , Retinol-Binding Proteins/metabolism , Vitamin A/metabolism , Animals , Female , Homeostasis , In Situ Hybridization , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Knockout , Retinol-Binding Proteins/deficiency , Retinol-Binding Proteins, Cellular , Vitamin A/administration & dosage , Vitamin A Deficiency/metabolism , Vitamin A Deficiency/pathology
11.
Dev Biol ; 208(1): 30-43, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10075839

ABSTRACT

We previously reported that mice lacking the RARgamma gene and one or both alleles of the RARbeta gene (i.e., RARbeta+/-/RARgamma-/- and RARbeta-/-/RARgamma-/- mutants) display a severe and fully penetrant interdigital webbing (soft tissue syndactyly), caused by the persistence of the fetal interdigital mesenchyme (Ghyselinck et al., 1997, Int. J. Dev. Biol. 41, 425-447). In the present study, these compound mutants were used to investigate the cellular and molecular mechanisms involved in retinoic acid (RA)-dependent formation of the interdigital necrotic zones (INZs). The mutant INZs show a marked decrease in the number of apoptotic cells accompanied by an increase of cell proliferation. This marked decrease was not paralleled by a reduction of the number of macrophages, indicating that the chemotactic cues which normally attract these cells into the INZs were not affected. The expression of a number of genes known to be involved in the establishment of the INZs, the patterning of the autopod, and/or the initiation of apoptosis was also unaffected. These genes included BMP-2, BMP-4, Msx-1, Msx-2, 5' members of Hox complexes, Bcl2, Bax, and p53. In contrast, the mutant INZs displayed a specific, graded, down-regulation of tissue transglutaminase (tTG) promoter activity and of stromelysin-3 expression upon the removal of one or both alleles of the RARbeta gene from the RARgamma null genetic background. As retinoic acid response elements are present in the promoter regions of both tTG and stromelysin-3 genes, we propose that RA might increase the amount of cell death in the INZs through a direct modulation of tTG expression and that it also contributes to the process of tissue remodeling, which accompanies cell death, through an up-regulation of stromelysin-3 expression in the INZs. Approximately 10% of the RARbeta-/- /RARgamma-/- mutants displayed a supernumerary preaxial digit on hindfeet, which is also a feature of the BMP-7 null phenotype (Dudley et al., 1995, Genes Dev. 9, 2795-2807; Luo et al., 1995, Genes Dev. 9, 2808-2820). BMP-7 was globally down-regulated at an early stage in the autopods of these RAR double null mutants, prior to the appearance of the digital rays. Therefore, RA may exert some of its effects on anteroposterior autopod patterning through controlling BMP-7 expression.


Subject(s)
Bone Morphogenetic Proteins/genetics , Receptors, Retinoic Acid/genetics , Transforming Growth Factor beta , Tretinoin/metabolism , Animals , Apoptosis/genetics , Bone Morphogenetic Protein 7 , Cell Division/genetics , Down-Regulation , Embryonic and Fetal Development , Forelimb/embryology , Forelimb/growth & development , Gene Expression Regulation, Developmental/genetics , Macrophages/metabolism , Matrix Metalloproteinase 11 , Metalloendopeptidases/genetics , Mice , Mice, Transgenic , Mutation , Transglutaminases/genetics , Retinoic Acid Receptor gamma
12.
Development ; 125(23): 4691-707, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9806918

ABSTRACT

We have engineered a mouse mutation that specifically deletes the C-terminal 18 amino acid sequence of the RXRalpha protein. This deletion corresponds to the last helical alpha structure (H12) of the ligand-binding domain (LBD), and includes the core of the Activating Domain of the Activation Function 2 (AF-2 AD core) that is thought to be crucial in mediating ligand-dependent transactivation by RXRalpha. The homozygous mutants (RXRalpha af2(o)), which die during the late fetal period or at birth, exhibit a subset of the abnormalities previously observed in RXRalpha -/- mutants, often with incomplete penetrance. In marked contrast, RXRalpha af2(o)/RXRbeta -/- and RXRalpha af2(o)/RXRbeta -/- /RXRgamma -/- compound mutants display a large array of malformations, which nearly recapitulate the full spectrum of the defects that characterize the fetal vitamin A-deficiency (VAD) syndrome and were previously found in RAR single and compound mutants, as well as in RXRalpha/RAR(alpha, beta or gamma) compound mutants. Analysis of RXRalpha af2(o)/RAR(alpha, beta or gamma) compound mutants also revealed that they exhibit many of the defects observed in the corresponding RXR alpha/RAR compound mutants. Together, these results demonstrate the importance of the integrity of RXR AF-2 for the developmental functions mediated by RAR/RXR heterodimers, and hence suggest that RXR ligand-dependent transactivation is instrumental in retinoid signalling during development.


Subject(s)
Abnormalities, Multiple/genetics , Embryonic and Fetal Development/genetics , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Sequence Deletion , Transcription Factors/genetics , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Animals, Newborn , Base Sequence , DNA Primers , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Death , Female , Fetal Death/genetics , Genomic Library , Homozygote , Ligands , Mice , Mice, Knockout , Molecular Sequence Data , Mutagenesis, Site-Directed , Pregnancy , Protein Structure, Secondary , Receptors, Retinoic Acid/chemistry , Recombinant Proteins/metabolism , Restriction Mapping , Retinoid X Receptors , Transcription Factors/chemistry , Transcriptional Activation , Vitamin D Deficiency/embryology , Vitamin D Deficiency/genetics
13.
J Histochem Cytochem ; 46(10): 1103-11, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9742066

ABSTRACT

Apart from the retinoic acid nuclear receptor family, there are two low molecular weight (15 kD) cellular retinoic acid binding proteins, named CRABPI and II. Mouse monoclonal and rabbit polyclonal antibodies were raised against these proteins by using as antigens either synthetic peptides corresponding to amino acid sequences unique to CRABPI or CRABPII, or purified CRABP proteins expressed in E. coli. Antibodies specific for mouse and/or human CRABPI and CRABPII were obtained and characterized by immunocytochemistry and immunoblotting. They allowed the detection not only of CRABPI but also of CRABPII in both nuclear and cytosolic extracts from transfected COS-1 cells, mouse embryos, and various cell lines.


Subject(s)
Cell Nucleus/chemistry , Receptors, Retinoic Acid/immunology , Animals , Antibodies/chemistry , COS Cells , Embryo, Mammalian/chemistry , Humans , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Receptors, Retinoic Acid/analysis , Transfection , Tumor Cells, Cultured
14.
Dev Biol ; 198(2): 303-18, 1998 Jun 15.
Article in English | MEDLINE | ID: mdl-9659935

ABSTRACT

To investigate the relative contribution of retinoic acid receptor (RAR)beta isoforms in conotruncal septation, RAR beta 1 and beta 3 were inactivated in the mouse. Mice lacking RAR beta 1 and beta 3 appear normal. Disruption of these isoforms in RAR alpha or RAR gamma null genetic backgrounds results in a high postpartum lethality. However, except for ocular defects found in RAR beta 1-3/RAR gamma compound mutants, the double null mutants display only abnormalities seen in single null mutants. This probably reflects a functional redundancy with other RARs, most notably with RAR beta 2 which is five- to sixfold more abundant than RAR beta 1 and beta 3 and whose domain of expression is largely overlapping. The conotruncal ridges form normally in retinoid X receptor (RXR)alpha/RAR beta compound mutants but fail to fuse, apparently as a result of excessive apoptosis of mesenchymal cells. Additionally, many cardiomyocytes in the conotruncal wall of these mutants appear necrotic. Although RAR beta 1 and beta 3 are expressed specifically in the conotruncal ridges, failure of fusion of these structures is not more frequent in RXR alpha/RAR beta 1-3 double null mutants than in RXR alpha single null mutants. Similarly, the disruption of the sole RAR beta 2 isoform in a RXR alpha null genetic background does not result in an increase of the frequency of conotruncal septum agenesis. However, this agenesis is fully penetrant in RXR alpha/RAR beta +/- mutants, which reflects distinct role of RXR alpha:RAR beta 1 (and beta 3) and RXR alpha:RAR beta 2 heterodimers in promoting the survival of conotruncal mesenchymal cells. Unexpectedly, we discovered that, in wild-type embryos, the conotruncal mesenchyme is a major site of morphogenetic cell death and that conotruncal myocytes are occasionally necrotic. Thus, excessive cell death in the conotruncus is a potential cause of ventricular septal defects in humans.


Subject(s)
Heart Septum/embryology , Receptors, Retinoic Acid/physiology , Animals , Cell Death , Female , Heart Septal Defects/embryology , Mice , Mice, Inbred C57BL , Morphogenesis , Pregnancy , Receptors, Retinoic Acid/genetics , Tretinoin/pharmacology
15.
Eur J Oral Sci ; 106 Suppl 1: 24-31, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9541199

ABSTRACT

The RAR and RXR families of retinoid nuclear receptors each comprise three isotypes (alpha, beta and gamma). In vitro, RARs bind to their cognate DNA response elements as heterodimers with RXRs. Null mutations of all six isotypes have been generated. The defects displayed by RAR alpha, beta and gamma single null mutant mice are confined to a small subset of the tissues normally expressing these receptors. This discrepancy reflects the existence of a functional redundancy, since RAR double null mutants exhibit congenital malformations in almost every organ system. In particular, most of the structures derived from the mesectoderm are severely affected. Analysis of mutant mice lacking both RARs and RXRs indicates that RXR alpha:RAR gamma heterodimers are instrumental in the patterning of craniofacial skeletal elements, whereas RXR alpha:RAR alpha heterodimers may be preferentially involved in the generation of neural crest cell-derived arterial smooth muscle cells. Both RXR alpha:RAR beta and RXR alpha:RAR gamma heterodimers appear to function during the development of the ocular mesenchyme. Moreover, atavistic reptilian cranial structures are generated in RAR mutants, suggesting that the RA signal has been implicated in the modification of developmental programs in the mesectoderm during evolution.


Subject(s)
Ectoderm/metabolism , Mesoderm/metabolism , Tretinoin/metabolism , Abnormalities, Multiple/embryology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Animals , Biological Evolution , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Dimerization , Eye/embryology , Eye/metabolism , Facial Bones/embryology , Facial Bones/metabolism , Mice , Mice, Mutant Strains , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Odontogenesis/genetics , Odontogenesis/physiology , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors , Skull/embryology , Skull/metabolism , Thymus Gland/embryology , Thymus Gland/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Int J Dev Biol ; 41(3): 425-47, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9240560

ABSTRACT

Homozygous RAR beta mutants are growth-deficient, but are fertile and have a normal longevity. They display homeotic transformations and malformations of cervical vertebrae and a retrolenticular membrane. This latter abnormality arises from the persistence and hyperplasia of the primary vitreous body. In contrast, we found that abnormalities of cranial nerves IX and X which were previously proposed to be specific features of the RAR beta mutant phenotype (Luo et al., Mech. Dev. 53: 61-71, 1995) occur with the same low penetrance in wildtype littermates. Although the RAR beta protein is expressed at high levels in the striatum and interdigital mesenchyme, the brain and limbs of RAR beta mutants appear morphologically normal. RAR alpha/RAR beta double mutants display numerous visceral abnormalities, most of which are incompatible with post-natal life. The majority of these abnormalities was previously detected in RAR alpha/RAR beta2 mutants with the notable exceptions of agenesis of the stapedial (2nd aortic arch-derived) artery, thymic and spleen agenesis and abnormal inferior vena cava. RAR beta/RAR gamma double mutants show major ocular defects including a shortening of the ventral retina and pre-natal retinal dysplasia, both of which represent the only abnormalities of the fetal vitamin-A deficiency (VAD) syndrome not previously detected in RAR beta2/RAR gamma compound mutants. In addition, RAR beta is apparently functionally redundant with either RAR alpha or RAR gamma for the formation of a small subset of craniofacial skeletal elements, as well as for eyelid development and digit separation. We also provide evidence that, at least in some instances, this phenomenon of functional redundancy between RARs may be an artifactual consequence of gene knock-out.


Subject(s)
Embryonic and Fetal Development/physiology , Eye Abnormalities/genetics , Gene Expression Regulation, Developmental , Receptors, Retinoic Acid/physiology , Animals , Body Weight , Bone and Bones/abnormalities , Bone and Bones/pathology , Eye/embryology , Eye/growth & development , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Female , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Morphogenesis , Pregnancy , Receptors, Retinoic Acid/genetics , Thymus Gland/abnormalities , Thymus Gland/pathology
17.
C R Seances Soc Biol Fil ; 191(1): 77-90, 1997.
Article in French | MEDLINE | ID: mdl-9181129

ABSTRACT

Two families of nuclear receptors for retinoic acid (RA) have been characterized. Members of the RAR family (types alpha, beta and gamma and their isoforms alpha 1, alpha 2, beta 1 to beta 4, and gamma 1 and gamma 2) are activated by most physiologically occurring retinoids (all-trans RA, 9-cis RA, 4oxo RA and 3,4 dihyroRA). In contrast, members of the RXR family (types alpha, beta and gamma and their isoforms) are activated by 9cis-RA only. In addition to the multiplicity of receptors, the complexity of retinoid signalling is further increased by the fact that, at least in vitro, RARs bind to their cognate response elements as heterodimers with RXRs. Moreover, RXRs can also bind, in vitro, to some DNA elements as homodimers, and are heterodimeric partners for other nuclear receptors, including TRs, VDR, PPARs and a number of orphan nuclear receptors. To evaluate the functions of the different RARs and RXRs types and isoforms, we have generated null mutant mice by targeted gene disruption in ES cells. As to the functions of RARs, we found that RAR alpha 1 and RAR gamma 2 null mutant mice are apparently normal. Mice deficient in RAR alpha or RAR gamma (i.e., all alpha or gamma isoforms disrupted) show aspects of the post-natal vitamin A deficiency (VAD) syndrome which can be cured or prevented by RA, including post-natal lethality, poor weight gain and male sterility. RAR beta 2 (and RAR beta) null mutants display a retrolenticular membrane which represents the most frequent defect of the fetal VAD syndrome. That these abnormalities were restricted to a small subset of the tissues normally expressing these receptors suggested that some degree of functional redundancy should exist in the RAR family. To test this hypothesis we then generated RAR double null mutants. RAR alpha beta, RAR alpha gamma and RAR beta gamma compound mutants exhibit all the malformations of the fetal VAD syndrome, thus demonstrating that RA is the vitamin A derivative which plays a crucial role at many different stages and in different structures during organogenesis. Interestingly, almost all the structures derived from mesenchymal neural crests cells (NCC) are affected in RAR compound mutants. As to the functions of RXRs, RXR gamma null mutants are viable, fertile and morphologically normal. In contrast, RXR alpha null fetuses display a thin ventricular wall and die in utero from cardiac failure. A myocardial hypoplasia has also been observed in some RAR compound mutants as well as in VAD fetuses. Thus, RXR alpha seems to act as an inhibitor of ventricular cardiocyte differentiation and/or as a positive regulator of their proliferation, and these functions might involve heterodimerization with RARs and activation by RA. RXR beta null mutants are viable but the males are sterile, most probably because of an abnormal lipid metabolism in the Sertoli cells. New abnormalities, absent in RXR alpha mutants, are generated in RXR alpha/RAR (alpha, beta or gamma) compound mutants. All these abnormalities are also seen in RAR double mutants as well as in VAD fetuses. In contrast, such manifestations of synergism are not observed between the RXR beta or RXR gamma and the RAR (alpha, beta or gamma) null mutations. These data strongly support the conclusion that RXR alpha/RAR heterodimers represent the main functional units of the RA signalling pathway during embryonic development. Moreover, since RXR gamma-/-/RXR beta-/-/RXR alpha +/-mutants are viable, a single allele of RXR alpha can perform most of the developmental RXR functions.


Subject(s)
Embryonic and Fetal Development/genetics , Tretinoin/physiology , Animals , Dimerization , Mice , Mice, Mutant Strains , Phenotype , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/physiology
18.
Mol Endocrinol ; 7(2): 258-72, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8469239

ABSTRACT

Genomic clones containing the gene for the glutathione peroxidase-like androgen-regulated murine epididymal protein of 24 kilodaltons (arMEP24) were isolated. A 9-kilobase DNA fragment was sequenced and found to contain the entire coding region of the gene, which is divided into five exons. The exact sizes and boundaries of the exon blocks were deduced by comparison with the cDNA sequence. One major and four weak transcription initiation sites in the epididymis were localized by primer extension. The promoter of the gene does not contain a conventional TATA box immediately up-stream of the start site; rather, the sequence TATCA occurs at residue -35. Two CAAT boxes in opposite orientation and two putative binding sites for the transcription factor Sp1 were identified up-stream of the TATA-like box. To localize the cis-acting sequences responsible for androgen regulation of expression, fragments of the arMEP24 gene promoter region were cloned in front of the luciferase (LUC) reporter gene and cotransfected with an androgen receptor expression vector into CV-1 cells in a transient assay. LUC activities of CV-1 cells grown in the presence of various concentrations of 5 alpha-dihydrotestosterone were compared to LUC activities of untreated controls. The DNA fragment containing up to 200 nucleotides up-stream from the major transcription start site was sufficient for the full promoter activity, but not for the responsiveness to androgen induction. Depending on the 5 alpha-dihydrotestosterone concentration, a 2- to 4-fold induction of LUC activity was found if a -1797 to -167 arMEP24 gene fragment was used linked to the reporter gene driven by either the homologous promoter or the heterologous thymidine kinase promoter. Two or three copies of the imperfect palindromic sequence TGTTGAgagAGAACA, found at position -896 to -882 in the gene and resembling the consensus steroid hormone-responsive element, are able to confer androgen regulation to the thymidine kinase promoter independently of their orientation. These findings support evidence that transcriptional regulation of the arMEP24 gene occurs via the sequence TGTTGAgagAGAACA. Homologies found in the sequence up-stream of the promoter with several putative binding sites for erythroid-specific trans-acting regulatory proteins are discussed. Finally, the arMEP24 gene is located by in situ hybridization in the [A2-A4] region of mouse chromosome 13.


Subject(s)
Epididymis/enzymology , Glutathione Peroxidase , Testicular Hormones/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Consensus Sequence , Dihydrotestosterone/pharmacology , Gene Expression Regulation, Enzymologic , Genes , Male , Mice , Mice, Inbred BALB C/genetics , Molecular Sequence Data , Promoter Regions, Genetic , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Transcription, Genetic
19.
Mol Cell Endocrinol ; 89(1-2): 67-77, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1301385

ABSTRACT

The protein MEP24 was previously described as a glutathione peroxidase-like molecule specifically secreted by the mouse caput epididymidis. Recently, its binding to the head of spermatozoa was demonstrated. Here, the regulation of MEP24 expression was studied by analyzing transcriptional and translational activities in the epididymis (1) of adult mice castrated on day 60 and given various substitutive testosterone (T) treatments from day 90 and (2) of hemicastrated adult animals. In castrated mice, T treatment induced a significant rise in plasma T and 5 alpha-dihydrotestosterone (DHT) concentrations that greatly exceeded the control values. Owing to efficient regulation, however, the epididymal T and DHT levels were never higher than those of the controls. The restoration of MEP24 mRNA accumulation was complete when the epididymal DHT content returned to its normal value. However, when estimated in a cell-free system, the in vitro translatable MEP24 mRNA level never exceeded 70% of control values, even though the DHT and accumulated mRNAs were restored by 100% or more. In hemicastrates, the T content was normal on the castrated side, while the DHT content exhibited a significant decrease (47%). In this case, the MEP24 mRNA accumulation reached 88% of the normal value, but the translation rate, both in vitro and in vivo, was only about 50%. Ultrastructural studies showed that the normal rough endoplasmic reticulum organization in segment I cells is dependent upon the presence of testicular fluid in the epididymal duct lumen. Thus, this report shows that the MEP24 mRNA steady-state level is completely recovered in the presence of a normal epididymal DHT content, while restoration of the regulation of translation is just partial. This could be related to the cell organization but seems mainly dependent upon the presence of specific mRNA-associated factors which are probably under the control of androgens and/or molecules carried by the testicular fluid.


Subject(s)
Epididymis/drug effects , Gene Expression Regulation/drug effects , Glutathione Peroxidase , Protein Biosynthesis , Testicular Hormones , Testosterone/analogs & derivatives , Testosterone/pharmacology , Animals , Epididymis/chemistry , Epididymis/metabolism , Epididymis/ultrastructure , Epithelium/metabolism , Epithelium/ultrastructure , Male , Mice , Orchiectomy , Protein Biosynthesis/drug effects , Proteins/genetics , RNA, Messenger/biosynthesis , Testosterone/analysis , Transcription, Genetic/drug effects
20.
J Endocrinol ; 133(2): 197-203, 1992 May.
Article in English | MEDLINE | ID: mdl-1613422

ABSTRACT

The pattern of labelled proteins synthesized and secreted in vitro by the adult mouse epididymis was studied by one- and two-dimensional polyacrylamide gel electrophoresis. The presence of a major 24 kDa protein synthesized and secreted in a tissue-specific manner by the caput epididymidis was detected. For this molecular weight, two-dimensional analysis indicated several proteins including two polypeptides (pI 8.4 and 8.8) whose expression is under androgenic control. Partial amino acid sequence analysis showed a complete N-terminal identity between these two peptides. A polyclonal monospecific immune serum was raised against the two proteins. Only purified immunoglobulins precipitated them, showing that immunological affinity is restricted to these two proteins in the epididymis. Indirect immunofluorescence assay revealed specific binding of antibodies on the acrosomal region of spermatozoa isolated from the caput, corpus or cauda epididymides. Testicular spermatozoa were not labelled under the same conditions. To investigate the physiological role of androgens in the synthesis and secretion of the 24 kDa proteins, tissue slices of epididymides from adult mice which had been castrated, or castrated then injected with testosterone were incubated with [35S]methionine. Castration and testosterone replacement kinetics revealed that alterations in 24 kDa protein synthesis follow immediately upon androgenic privation and replacement.


Subject(s)
Epididymis/metabolism , Protein Biosynthesis , Testosterone/physiology , Animals , Culture Techniques , Electrophoresis, Polyacrylamide Gel , Male , Mice , Molecular Weight , Proteins/chemistry , Spermatozoa/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...