Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Sex Dev ; 6(5): 223-30, 2012.
Article in English | MEDLINE | ID: mdl-22797524

ABSTRACT

In an effort to identify novel candidate genes involved in testis determination, we previously used suppression subtraction hybridisation PCR on male and female whole embryonic (12.0-12.5 days post coitum) mouse gonads. One gene to emerge from our screen was Redd1. In the current study, we demonstrate by whole-mount in situ hybridisation that Redd1 is differentially expressed in the developing mouse gonad at the time of sex determination, with higher expression in testis than ovary. Furthermore, Redd1 expression was first detected as Sry expression peaks, immediately prior to morphological sex determination, suggesting a potential role for Redd1 during testis development. To determine the functional importance of this gene during testis development, we generated Redd1-deficient mice. Morphologically, Redd1-deficient mice were indistinguishable from control littermates and showed normal fertility. Our results show that Redd1 alone is not required for testis development or fertility in mice. The lack of a male reproductive phenotype in Redd1 mice may be due to functional compensation by the related gene Redd2.


Subject(s)
Reproduction/physiology , Testis/embryology , Transcription Factors/metabolism , Animals , Biomarkers/metabolism , Crosses, Genetic , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Female , Fertility , Fetus/embryology , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental , Male , Mice , Phenotype , Polymerase Chain Reaction , Reproducibility of Results , Sertoli Cells/metabolism , Testis/cytology , Testis/growth & development , Testis/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics
2.
Sex Dev ; 5(1): 7-15, 2011.
Article in English | MEDLINE | ID: mdl-21099207

ABSTRACT

Mammalian sex determination is a dynamic process involving balanced gene expression leading to the development of either a testis or an ovary. Candidate sex-determining genes have been identified through microarray-based studies of gonadal gene expression; however, few methods exist for validation. This study describes a new technique for transfecting gonads using nucleofection. Fifteen micrograms of expression plasmid DNA was transfected into E11.5 gonads, cultured for 3 days and gene expression analyzed. Following optimization, we consistently achieved cell transfection efficiencies of 11% of cells using pMax-GFP plasmid. To test the applicability of nucleofection to studies of gene function, a testis-determining gene was transfected into gonads and its ability to sex-reverse was examined. When Sry was transfected into female (XX) gonads, upregulation of its target gene Sox9 was observed, as well as a downregulation of the ovarian gene Foxl2. Conversely, when shSox9 was introduced into male (XY) gonads, reduction of Sox9 and its target gene, Amh was observed, with a concomitant upregulation of Foxl2. Nucleofection-based gene delivery can recapitulate in vivo events of gonadal development that demonstrates 'proof-of-principle' of the method as a screening tool to evaluate the cellular function of potential sex-determining and gonadal differentiation genes.


Subject(s)
Ovary/embryology , Ovary/metabolism , Sex Determination Processes/genetics , Sex Differentiation/genetics , Testis/embryology , Testis/metabolism , Animals , Embryo Culture Techniques , Female , Forkhead Box Protein L2 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genes, sry , Green Fluorescent Proteins/genetics , Male , Mice , Recombinant Fusion Proteins/genetics , SOX9 Transcription Factor/antagonists & inhibitors , SOX9 Transcription Factor/genetics , Sex-Determining Region Y Protein/genetics , Transfection
3.
Sex Dev ; 4(6): 321-5, 2010.
Article in English | MEDLINE | ID: mdl-20838034

ABSTRACT

During mouse sex determination, SRY upregulates the core testis-specific enhancer of Sox9, TESCO. Mutations in human SRY are found in one third of cases with XY pure gonadal dysgenesis (XY GD; Swyer syndrome), while two thirds remain unexplained. Heterozygous SOX9 mutations can cause XY GD in association with the skeletal malformation syndrome campomelic dysplasia. We hypothesized that human TESCO mutations could cause isolated XY GD. Sixty-six XY GD cases with an intact SRY were analyzed for TESCO point mutations or deletions. No mutations were identified. We conclude that TESCO mutations are not a common cause of XY GD.


Subject(s)
Gonadal Dysgenesis, 46,XY/genetics , Homeodomain Proteins/genetics , Mutation/genetics , Sex-Determining Region Y Protein/genetics , Tumor Suppressor Proteins/genetics , Animals , Cytoskeletal Proteins , Humans , LIM Domain Proteins , Male , Mice , RNA-Binding Proteins
4.
Sex Dev ; 3(4): 194-204, 2009.
Article in English | MEDLINE | ID: mdl-19752599

ABSTRACT

In biological research, quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) assays are commonly employed to study mRNA abundance in cells and tissues. This type of assay usually relies on assessing transcript abundance relative to constitutively expressed endogenous reference genes. Therefore, it is important that the reference genes themselves are stably expressed in the cells or tissues analyzed, independent of factors such as age, sex, disease or experimental manipulations. Since no gene is expressed at the same level in all cells at all times, suitable reference genes must be identified for the specific cellular system or tissue being investigated. Here, we sought to identify stably expressed endogenous reference genes during embryonic gonad development in the mouse. We measured the transcript abundance of 10 frequently employed normalizing genes, of which 4 were stably expressed in fetal gonads from 11.5 to 14.5 dpc irrespective of sex. Based on our analysis, we suggest that Rn18s, Rps29, Tbp and Sdha are suitable reference genes for qRT-PCR expression studies during early gonad differentiation in the mouse.


Subject(s)
Fetus/metabolism , Gene Expression Regulation, Developmental , Gonads/embryology , Gonads/metabolism , Reverse Transcriptase Polymerase Chain Reaction/standards , Animals , DNA Primers/analysis , Electrophoresis, Agar Gel , Gene Expression Profiling , Mesonephros/embryology , Mesonephros/metabolism , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reference Standards
5.
Sex Dev ; 1(2): 114-26, 2007.
Article in English | MEDLINE | ID: mdl-18391522

ABSTRACT

The study of the mammalian sex-determining pathway has been hampered by the lack of cell culture systems to investigate the underlying molecular relationships between sex-determining genes. Recent approaches using high-throughput genome-wide studies have revealed a number of sexually dimorphic genes expressed in the developing mouse gonad. Here, we investigated a human testicular cell line in terms of its expression of known sex-determining genes and newly identified candidates. The human embryonal carcinoma cell line NT2/D1 was screened for the expression of 46 genes with known or potential roles in the sex-determining and differentiation pathway. Forty genes tested were expressed in NT2/D1 cells including the testis-determining genes SRY, SOX9, SF-1, DHH and FGF9. Genes not expressed included WT1, DAX1 and the ovary-specific genes FOXL2 and WNT4. Cell-specific markers demonstrate that NT2/D1 cells reflect a number of cell types in the gonad including Sertoli, Leydig and germ cells. Our results suggest that male pathways initiated by SRY, SOX9 and SF-1 remain intact in these cells. Lack of expression of ovary-specific genes is consistent with a commitment of these cells to the male lineage. Manipulation of gene expression in this cell line could be an important new in vitro tool for the discovery of new human sex-determining genes.


Subject(s)
Gene Expression Profiling , Urogenital System/metabolism , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Germ Cells/metabolism , Gonads/metabolism , Humans , Leydig Cells/metabolism , Male , Ovary/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sertoli Cells/metabolism , Sex Determination Processes , Sex Differentiation/genetics , Signal Transduction/genetics , Testis/metabolism , Urogenital System/embryology
6.
Cytogenet Genome Res ; 101(3-4): 185-98, 2003.
Article in English | MEDLINE | ID: mdl-14684982

ABSTRACT

The decision of the bi-potential gonad to develop into either a testis or ovary is determined by the presence or absence of the Sex-determining Region gene on the Y chromosome (SRY). Since its discovery, almost 13 years ago, the molecular role that SRY plays in initiating the male sexual development cascade has proven difficult to ascertain. While biochemical studies of clinical mutants and mouse genetic models have helped in our understanding of SRY function, no direct downstream targets of SRY have yet been identified. There are, however, a number of other genes of equal importance in determining sexual phenotype, expressed before and after expression of SRY. Of these, one has proven of central importance to mammals and vertebrates, SOX9. This review describes our current knowledge of SRY and SOX9 structure and function in the light of recent key developments.


Subject(s)
DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Mammals/genetics , Nuclear Proteins , Sex Determination Processes , Transcription Factors/physiology , Animals , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Genes, sry , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/genetics , Humans , Male , Mice , Models, Genetic , Mutation , SOX9 Transcription Factor , Sex-Determining Region Y Protein , Transcription Factors/chemistry , Transcription Factors/genetics
7.
Cytogenet Genome Res ; 101(3-4): 219-23, 2003.
Article in English | MEDLINE | ID: mdl-14684986

ABSTRACT

SOX9 is expressed at the onset of the genital ridge formation in both sexes. It is assumed that SRY, the testis determining gene, turns SOX9 on in male embryos because it is turned off in female embryos. Spatial expression of SRY follows a cranio-caudal pattern. Here, we asked if SOX9 is expressed in the same cell lineage and with a similar pattern as SRY. A correlative study between the structural changes in the genital ridge and the immunocytochemical localization of SOX9-positive cells was undertaken. We used a transgenic strain expressing the green fluorescent protein (GFP) that considerably enhanced the cell context where the first SOX9-positive cells appear. Although SOX9-positive cells are located among loose mesenchymal cells by stages of 8-14 tail somites (ts) in both sexes, they are absent in the thickening coelomic epithelium of females. At 15 ts the first SOX9-positive cells appear within the core of the condensed cells only in male genital ridges. At 17 ts, a gradient of SOX9-positive cells in males is apparent, closely following the cranio-caudal pattern of cell aggregation seen in genital ridges of both sexes. Hence, our results suggest that SOX9 is expressed only in loose mesenchymal cells in both sexes and that expression of SOX9 in males requires the prior aggregation of cells in the genital ridges. The correspondence of SOX9 and SRY pattern of expression supports that both genes are expressed in the preSertoli cell lineage in the core of the genital ridges.


Subject(s)
Genitalia/cytology , High Mobility Group Proteins/metabolism , Sertoli Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Aggregation , Cell Lineage , Female , Genitalia/embryology , Genitalia/metabolism , Green Fluorescent Proteins , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Male , Mice , Mice, Transgenic , SOX9 Transcription Factor , Stem Cells/cytology , Stem Cells/physiology
8.
J Dent Res ; 82(9): 725-30, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12939358

ABSTRACT

Regulatory factors governing the formation of bone in the glenoid fossa in response to functional appliance therapy have not been identified. Therefore, the purpose of this study was to investigate the temporal pattern of expression of two key chondrogenesis markers-SOX9 and its target gene, type II collagen-in the glenoid fossa by immunostaining in a 35-day-old Sprague Dawley rat model during both natural growth and forward mandibular positioning. The expression of both factors was up-regulated when the mandible was positioned forward, indicating an enhancement of chondrocyte differentiation and chondroid matrix formation. Our results indicate that chondroid bone formation in the glenoid fossa in response to forward mandibular positioning is regulated by molecular markers indicative of endochondral ossification.


Subject(s)
Collagen Type II/analysis , High Mobility Group Proteins/analysis , Mandible/anatomy & histology , Orthodontic Appliances, Functional , Temporal Bone/metabolism , Transcription Factors/analysis , Up-Regulation , Analysis of Variance , Animals , Cell Differentiation/drug effects , Chondrocytes/drug effects , Chondrogenesis/drug effects , Collagen Type II/genetics , Extracellular Matrix/drug effects , Female , High Mobility Group Proteins/genetics , Mandible/growth & development , Models, Animal , Osteogenesis/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , SOX9 Transcription Factor , Temporal Bone/growth & development , Transcription Factors/genetics , Up-Regulation/genetics
9.
Appl Occup Environ Hyg ; 18(8): 597-619, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12851010

ABSTRACT

During the summer of 1994, football players at a practice field reported noxious odors in the area. Ohio Environmental Protection Agency (OEPA) investigations of industries surrounding the field included a printing facility producing vinyl shower curtains with screen-printed designs. Though not the source of the odor, they were discharging volatile organic compounds directly to the environs in violation of OEPA regulations. To achieve compliance they installed a catalytic oxidizer for treating discharged air. Due to high equipment costs, the capacity of the installed catalytic oxidizer resulted in a substantial reduction in discharged air flow rates and increased solvent vapor concentrations within the workplace. Vapor levels caused worker discomfort, prompting a request for assistance from the Ohio Bureau of Workers Compensation. The vapor concentrations were found to exceed NIOSH, OSHA, and ACGIH acceptable exposure levels. The workers were then required to wear organic vapor removing respirators full-time while printing as a temporary protective measure. The company requested NIOSH assistance in finding methods to reduce solvent vapor concentrations. NIOSH studies included the identification of the sources and relative magnitude of solvent emissions from the printing process, the design of controls for the emissions, and the development of substitute inks using non-photochemically reactive solvents. The new ink system and controls allowed OEPA removal of the requirement for the treatment of discharged air and substantial increases in dilution ventilation. Increased ventilation would permit reduction in worker exposures to less than 1/3 mixture TLV levels and removal of requirements for respirator usage. This solution was the result of a comprehensive review of all facets of the problem, including OEPA regulations. It also required cooperative work between the company and federal, state, and local governmental agencies.


Subject(s)
Guideline Adherence , Ink , Occupational Exposure/prevention & control , Vinyl Compounds/chemistry , Workplace , Air Movements , Humans , Manufactured Materials , National Institute for Occupational Safety and Health, U.S. , Odorants , Ohio , United States , Ventilation , Volatilization
10.
Mol Endocrinol ; 17(7): 1332-43, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12677004

ABSTRACT

Chondrogenesis leads to the formation of mature cartilage and generates initial skeletal elements that serve as templates for endochondral bone formation. Bone morphogenetic proteins (BMPs) are involved in several developmental and organogenetic processes and have been identified as key regulators in chondrogenesis. In the present study we sought to determine the transcriptional mechanisms contributing to the induction of chondrogenic markers by BMP-2. Time-course studies with BMP-2-stimulated C3H10T1/2 cells showed a dose-dependent appearance of Alcian-blue-positive material and up-regulated expression of type-II collagen mRNA. This last effect required new protein synthesis because addition of cycloheximide completely blocked the induction of type-II collagen mRNA. A region encompassing the chondrocyte-specific enhancer, localized in intron I of type-II collagen alpha1 chain (Col2a1) gene, is sufficient to confer BMP-2-dependent transcriptional induction of type-II collagen gene expression. Analysis of the expression levels of chondrogenic Sry-type high-mobility group (HMG) box proteins (SOX) transcription factors demonstrated a time-dependent induction of Sox6 expression by BMP-2 that correlated with the appearance of BMP-2- induced protein complexes bound to the chondrocyte-specific enhancer. Preincubation of nuclear extracts with SOX6 and SOX9 antibodies markedly reduced the intensity of these bands. Forced expression of SOX6 mimicked the BMP-2 effect, whereas coexpression of SOX9 promoted a synergistic interaction between both factors in transcription from the chondrocyte-specific enhancer. Moreover, overexpression of a SOX6 mutated form, devoid of its high-mobility group domain, was sufficient to prevent transcriptional induction of the chondrocyte-specific enhancer by BMP-2. Taken together, these results indicate that SOX6 is an important downstream mediator of BMP-2 signaling in chondrogenesis.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Differentiation/physiology , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Nuclear Proteins , Transcription Factors/metabolism , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cell Line , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/physiology , Collagen Type II/drug effects , Collagen Type II/genetics , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic , Gene Expression Regulation , High Mobility Group Proteins/drug effects , High Mobility Group Proteins/genetics , Mesoderm/cytology , Mice , Mutation , Response Elements/drug effects , Response Elements/genetics , SOX9 Transcription Factor , SOXD Transcription Factors , Sex-Determining Region Y Protein , Signal Transduction , Transcription Factors/drug effects , Transcription Factors/genetics , Transcription, Genetic
11.
Cell Transplant ; 11(6): 499-505, 2002.
Article in English | MEDLINE | ID: mdl-12428738

ABSTRACT

The immunoprivileged environment of the testes was first described in the 1930s, and the Sertoli cell was later identified as the main cell type responsible for this phenomenon. Recent work has examined the possibility of recreating this immunoprivileged environment at heterotopic sites using isolated Sertoli cells. These studies have focused on protection of pancreatic islets and neuronal cells from immune destruction in the hopes of reversing type I diabetes and Parkinson's disease. The absence of a definitive marker for identifying Sertoli cells at the transplant site has been an obstacle to this research. The current study examines the presence of a nuclear transcription factor, Sox9, which is preferentially expressed in Sertoli cells. Syngeneic Lewis rat Sertoli cells were transplanted into the renal subcapsular space and a subcutaneous site in Lewis female rats and examined histologically 21 days later. In addition, porcine Sertoli cells were transplanted into the renal subcapsular space in female SCID mice. Control testes and the transplant sites were examined immunohistochemically using an antibody to Sox9. The results from the study demonstrate that Sox9 expression is restricted to the Sertoli cells of the neonatal rat and porcine testis, indicating high homology between species. In addition, Sox9 expression was also observed in the testicular-like tubules that formed in both syngeneic and xenogeneic heterotopic transplants in rats and SCID mice. The Sox9 expression was restricted to the regions where Sertoli cells would be found in the native testis. These results suggest that the Sox9 protein is a useful marker in identifying Sertoli cells in heterotopic transplants in a manner similar to insulin as a marker for pancreatic islets.


Subject(s)
High Mobility Group Proteins/analysis , Sertoli Cells/transplantation , Transcription Factors/analysis , Animals , Animals, Newborn , Biomarkers/analysis , Cell Transplantation/methods , Female , Male , Mice , Mice, SCID , Rats , Rats, Inbred Lew , SOX9 Transcription Factor , Sertoli Cells/physiology , Sex Differentiation , Swine
12.
Eur J Ophthalmol ; 12(6): 450-8, 2002.
Article in English | MEDLINE | ID: mdl-12510713

ABSTRACT

PURPOSE: To study the expression and distribution of transcription factor Sox9 and type IIA procollagen in the developing and aging eyes of normal and transgenic Dell mice carrying pro(alpha)1(II) collagen transgenes with a short deletion mutation, which cause ocular abnormalities in this mouse line. METHODS: The eyes of Del1 mice were studied on embryonic days E14.5, E16.5 and E18.5, and at the ages of 4 and nine months, using their nontransgenic littermates as controls. Sox9 and pro(alpha)1(IIA) collagen were detected by RNase protection assay and immunohistochemistry. RESULTS: RNase protection assay revealed Sox9 transcripts in the eyes of Del1 and control mice during development and aging. The mRNA for type IIA procollagen had a similar temporal expression pattern. On embryonic days E14.5, E16.5 and E18.5, Sox9 was located by immunohistochemistry in the nuclei and type IIA procollagen in the extracellular space of the developing retina. During growth and aging, the ocular expression of Sox9 mRNA and the immunohistochemical reaction for Sox9 antibody diminished, concomitant with the reduction in type II procollagen mRNA. However, at the age of nine months, levels of Sox9 and type IIA procollagen mRNAs were higher in the degenerating eyes of Del1 and control mice. CONCLUSIONS: The similarities in the temporo-spatial distribution of Sox9 and type IIA procollagen suggest that this transcription factor is involved in the activation of type II collagen expression in the eye, as has been demonstrated in prechondrogenic mesenchyme and immature cartilage. The increased production of Sox9 and type IIA procollagen in the aging retina and vitreous is analogous to degenerating articular cartilage where attempted tissue repair has also been observed.


Subject(s)
Aging/metabolism , Collagen Type II/genetics , High Mobility Group Proteins/metabolism , Mutation , Peptide Fragments/metabolism , Procollagen/metabolism , Retina/embryology , Transcription Factors/metabolism , Vitreous Body/embryology , Animals , Down-Regulation , High Mobility Group Proteins/genetics , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Peptide Fragments/genetics , Procollagen/genetics , RNA, Messenger/metabolism , Retina/growth & development , Retina/metabolism , SOX9 Transcription Factor , Transcription Factors/genetics , Vitreous Body/growth & development , Vitreous Body/metabolism
13.
J Bone Miner Res ; 16(10): 1837-45, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11585348

ABSTRACT

Fracture repair is the best-characterized situation in which activation of chondrogenesis takes place in an adult organism. To better understand the mechanisms that regulate chondrogenic differentiation of mesenchymal progenitor cells during fracture repair, we have investigated the participation of transcription factors L-Sox5, Sox6, and Sox9 in this process. Marked up-regulation of L-Sox5 and Sox9 messenger RNA (mRNA) and smaller changes in Sox6 mRNA levels were observed in RNAse protection assays during early stages of callus formation, followed by up-regulation of type II collagen production. During cartilage expansion, the colocalization of L-Sox5, Sox6, and Sox9 by immunohistochemistry and type II collagen transcripts by in situ hybridization confirmed a close relationship of these transcription factors with the chondrocyte phenotype and cartilage production. On chondrocyte hypertrophy, production of L-Sox5, Sox9 and type II collagen were down-regulated markedly and that of type X collagen was up-regulated. Finally, using adenovirus mediated bone morphogenetic protein 2 (BMP-2) gene transfer into fracture site we showed accelerated up-regulation of the genes for all three Sox proteins and type II collagen in fractures treated with BMP-2 when compared with control fractures. These data suggest that L-Sox5, Sox6, and Sox9 are involved in the activation and maintenance of chondrogenesis during fracture healing and that enhancement of chondrogenesis by BMP-2 is mediated via an L-Sox5/Sox6/Sox9-dependent pathway.


Subject(s)
Bone Morphogenetic Proteins/genetics , DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Transforming Growth Factor beta , Up-Regulation , Animals , Bone Morphogenetic Protein 2 , Bony Callus/metabolism , Collagen Type II/genetics , Collagen Type II/metabolism , Fracture Healing , Gene Transfer Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , RNA, Messenger/analysis , SOX9 Transcription Factor , SOXD Transcription Factors
14.
J Biol Chem ; 276(49): 46575-82, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11535586

ABSTRACT

The sex-determining factor SRY is a DNA-binding protein that diverts primordial gonads from the ovarian pathway toward male differentiation to form testes. It gains access to the nucleus through two distinct nuclear localization signals (NLSs) that flank the high mobility group (HMG) DNA-binding domain, but the mechanisms through which these NLSs operate have not been studied. In this study, we reconstitute the nuclear import of SRY in vitro, demonstrating a lack of requirement for exogenous factors for nuclear accumulation and a significant reduction in nuclear transport in the presence of antibodies to importin beta but not importin alpha. Using a range of quantitative binding assays including enzyme-linked immunosorbent assay, fluorescence polarization, and native gel mobility electrophoresis, we assess the binding of importins to SRY, demonstrating a high affinity recognition (in the low nm range) by Imp beta independent of Imp alpha. In assessing the contribution of each NLS, we found that the N-terminal NLS was recognized poorly by importins, whereas the C-terminal NLS was bound by importin beta with similar affinity to SRY. We also found that RanGTP, but not RanGDP, could dissociate the SRY-importin beta complex in solution using FP. We describe a novel double-fluorescent label DNA binding assay to demonstrate mutual exclusivity between importin beta recognition and DNA binding on the part of SRY, which may represent an alternative release mechanism upon nuclear entry. This study represents the first characterization of the nuclear import pathway for a HMG domain-containing protein. Importantly, it demonstrates for the first time that recognition of SRY by Imp beta is of comparable affinity to that with which Imp alpha/beta recognizes conventional NLS-containing substrates.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/physiology , Nuclear Localization Signals , Nuclear Proteins , Transcription Factors , beta Karyopherins/metabolism , Amino Acid Sequence , Animals , Base Sequence , DNA/metabolism , DNA Primers , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescence Polarization , Molecular Sequence Data , Protein Transport , Rats , Sex Determination Processes , Sex-Determining Region Y Protein
15.
J Cell Biochem Suppl ; Suppl 36: 71-8, 2001.
Article in English | MEDLINE | ID: mdl-11455572

ABSTRACT

SOX9 is a transcription factor that is expressed in chondrocytes and regulates expression of chondrocyte phenotype related genes. Expression of these genes is known to be suppressed by retinoic acid (RA). We, therefore, examined whether the Sox9 gene expression is regulated by RA in chondrocytes. RA treatment suppressed Sox9 mRNA expression in primary chondrocytes prepared from newborn mouse rib cartilage within 12 h and this suppression lasted at least up to 24 h. The RA suppression of Sox9 mRNA levels was dose-dependent starting at 0.5 microM with a maximum at 1 microM. Nuclear run-on assays revealed that RA reduced the rate of transcription of Sox9 gene. Finally, Western blot analysis indicated that RA suppressed SOX9 protein levels in these chondrocytes. Furthermore, overexpression of SOX9 reversed RA suppression of Col2a1 enhancer activity. These observations indicate that RA suppresses Sox9 gene expression in chondrocytes at least in part through transcriptional events. J. Cell. Biochem. Suppl. 36: 71-78, 2001.


Subject(s)
Chondrocytes/metabolism , High Mobility Group Proteins/metabolism , Transcription Factors/metabolism , Tretinoin/metabolism , Animals , Blotting, Northern , Blotting, Western , Cells, Cultured , Down-Regulation , Enhancer Elements, Genetic , High Mobility Group Proteins/genetics , Mice , Mice, Inbred ICR , RNA, Messenger/metabolism , SOX9 Transcription Factor , Transcription Factors/genetics , Transcription, Genetic , Tretinoin/pharmacology
16.
Brain Res ; 906(1-2): 143-8, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11430871

ABSTRACT

Human mutations in the transcription factor gene, SOX9, cause campomelic dysplasia (CD), a severe dwarfism associated with brain abnormalities including dilation of lateral ventricles, hypoplasia of the corpus callosum and cerebellum defects. To improve our understanding of how SOX9 contributes to the molecular genetic pathway of brain development we sought to investigate the distribution of SOX9 protein in rat and mouse brain. The regions of SOX9 expression identified in this study correlated with the sites of reported brain abnormalities in CD patients. SOX9 immunoreactivity was observed in nuclei of scattered cells throughout the brain, in the ependymal layer and cells of the choroid plexus. In the forebrain most SOX9-immunoreactive nuclei co-localised with the glial astrocyte marker S-100. In the cerebellum, SOX9 was observed mostly in cells surrounding Purkinje cells, which were identified, by electron microscopy, as Golgi epithelial cells, also known as Bergmann glia. Using SOX9 antibody as a marker for the precursors of Bergmann glia, we traced their origin during mouse development. At embryonic day (E)14.5 and E16.5, SOX9 immunoreactivity was present mainly in the primordial choroid plexus, and ventricular zone. By E18.5, SOX9 was observed in the granular cell and Purkinje cell layers but no labelling was detectable in the external granular layer. These results suggest that SOX9 immunoreactivity is a marker for Bergmann cells during development and favour the proposed origin of the secondary glial scaffold arising from Bergmann cells derived exclusively from the ventricular zone.


Subject(s)
Cell Differentiation/physiology , Cerebellum/embryology , Cerebellum/metabolism , High Mobility Group Proteins/metabolism , Nervous System Malformations/metabolism , Neuroglia/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Cerebellum/ultrastructure , Choroid Plexus/embryology , Choroid Plexus/metabolism , Choroid Plexus/ultrastructure , Ependyma/embryology , Ependyma/metabolism , Ependyma/ultrastructure , Female , Fetus , Immunohistochemistry , Male , Mice , Microscopy, Electron , Nervous System Malformations/pathology , Nervous System Malformations/physiopathology , Neuroglia/ultrastructure , Rats , SOX9 Transcription Factor , Stem Cells/ultrastructure
17.
FEBS Lett ; 496(2-3): 75-80, 2001 May 11.
Article in English | MEDLINE | ID: mdl-11356186

ABSTRACT

The campomelic dysplasia/autosomal sex reversal protein SOX9 is an important developmental transcription factor, required for correct bone and testis formation. Through in vitro and in vivo studies we have identified the heat shock protein HSP70 as an interacting partner for SOX9 in chondrocyte and testicular cell lines. HSP70 forms a ternary complex with DNA-bound SOX9. The interaction between HSP70 and SOX9 is ATP-independent and involves a highly conserved region of SOX9 hitherto of unknown function and the C-terminal region of HSP70. Our results implicate HSP70-SOX9 interactions in the assembly of multi-protein complexes during SOX9-mediated transcription.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , High Mobility Group Proteins/metabolism , Transcription Factors/metabolism , Adenosine Triphosphate/metabolism , Animals , Binding Sites , Blotting, Western , Bone and Bones/embryology , Cell Line , Chondrocytes/metabolism , Electrophoresis, Polyacrylamide Gel , Gene Deletion , Humans , Immunohistochemistry , Male , Mutagenesis , Precipitin Tests , Protein Binding , Rats , Recombinant Proteins/metabolism , SOX9 Transcription Factor , Sex Determination Processes , Testis/embryology , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured
18.
J Endocrinol ; 169(3): 573-9, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11375127

ABSTRACT

SOX9 is a transcription factor that activates type II procollagen (Col2a1) gene expression during chondrocyte differentiation. Glucocorticoids are also known to promote chondrocyte differentiation via unknown molecular mechanisms. We therefore investigated the effects of a synthetic glucocorticoid, dexamethasone (DEX), on Sox9 gene expression in chondrocytes prepared from rib cartilage of newborn mice. Sox9 mRNA was expressed at high levels in these chondrocytes. Treatment with DEX enhanced Sox9 mRNA expression within 24 h and this effect was observed at least up to 48 h. The effect of DEX was dose dependent, starting at 0.1 nM and maximal at 10 nM. The half life of Sox9 mRNA was approximately 45 min in the presence or absence of DEX. Western blot analysis revealed that DEX also enhanced the levels of SOX9 protein expression. Treatment with DEX enhanced Col2a1 mRNA expression in these chondrocytes and furthermore, DEX enhanced the activity of Col2-CAT (chloramphenicol acetyltransferase) construct containing a 1.6 kb intron fragment where chondrocyte-specific Sry/Sox- consensus sequence is located. The enhancing effect of DEX was specific to SOX9, as DEX did not alter the levels of Sox6 mRNA expression. These data suggest that DEX promotes chondrocyte differentiation through enhancement of SOX9.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chondrocytes/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , High Mobility Group Proteins/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Culture Techniques , Cell Differentiation/drug effects , Chondrocytes/cytology , Chondrocytes/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , High Mobility Group Proteins/genetics , Mice , Mice, Inbred ICR , Procollagen/genetics , RNA, Messenger/genetics , SOX9 Transcription Factor , Transcription Factors/genetics , Transcription, Genetic/drug effects
19.
Mol Genet Metab ; 72(4): 356-9, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11286511

ABSTRACT

SOX13 is a member of the SOX family of transcription factors that encodes the type 1 diabetes autoantigen, ICA12. The SOX13 gene maps at chromosome 1q31.3-32.1 near a region containing a susceptibility locus for type 1 diabetes. SOX13 was assessed as a candidate susceptibility gene. Analysis of the SOX13 gene identified a number of single nucleotide polymorphisms and a polymorphic CA dinucleotide repeat. Linkage and association studies indicate that SOX13 is unlikely to make a substantial contribution to type 1 diabetes susceptibility.


Subject(s)
Autoantigens/genetics , Diabetes Mellitus, Type 1/genetics , High Mobility Group Proteins/genetics , Chromosome Mapping , Chromosomes, Human, Pair 1/genetics , DNA/metabolism , Family , Gene Frequency , Genotype , Humans , Polymorphism, Single Nucleotide , SOXD Transcription Factors
20.
J Biol Chem ; 276(30): 27864-72, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11323423

ABSTRACT

Human mutations in the transcription factor SOX9 cause campomelic dysplasia/autosomal sex reversal. Here we identify and characterize two novel heterozygous mutations, F154L and A158T, that substitute conserved "hydrophobic core" amino acids of the high mobility group domain at positions thought to stabilize SOX9 conformation. Circular dichroism studies indicated that both mutations disrupt alpha-helicity within their high mobility group domain, whereas tertiary structure is essentially maintained as judged by fluorescence spectroscopy. In cultured cells, strictly nuclear localization was observed for wild type SOX9 and the F154L mutant; however, the A158T mutant showed a 2-fold reduction in nuclear import efficiency. Importin-beta was demonstrated to be the nuclear transport receptor recognized by SOX9, with both mutant proteins binding importin-beta with wild type affinity. Whereas DNA bending was unaffected, DNA binding was drastically reduced in both mutants (to 5% of wild type activity in F154L, 17% in A158T). Despite this large effect, transcriptional activation in cultured cells was only reduced to 26% in F154L and 62% in A158T of wild type activity, suggesting that a small loss of SOX9 transactivation activity could be sufficient to disrupt proper regulation of target genes during bone and testis formation. Thus, clinically relevant mutations of SOX9 affect protein structure leading to compound effects of reduced nuclear import and reduced DNA binding, the net effect being loss of transcriptional activation.


Subject(s)
Abnormalities, Multiple/genetics , Active Transport, Cell Nucleus/genetics , Bone and Bones/abnormalities , DNA/metabolism , Disorders of Sex Development , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Mutation , Point Mutation , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , Adult , Amino Acid Sequence , Animals , COS Cells , Cell Nucleus/metabolism , Cells, Cultured , Circular Dichroism , Electrophoresis, Polyacrylamide Gel , Female , Genes, Dominant , Heterozygote , High Mobility Group Proteins/chemistry , Humans , Immunohistochemistry , Infant, Newborn , Karyopherins , Karyotyping , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Nuclear Proteins/metabolism , Phenotype , Polymorphism, Single-Stranded Conformational , Protein Binding , Protein Conformation , Protein Denaturation , Protein Structure, Tertiary , SOX9 Transcription Factor , Sequence Analysis, DNA , Spectrometry, Fluorescence , Structure-Activity Relationship , Temperature , Transcription Factors/chemistry , Transfection , Tryptophan/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...