Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Prostate ; 79(4): 370-378, 2019 03.
Article in English | MEDLINE | ID: mdl-30488457

ABSTRACT

BACKGROUND: PSMA expression in the prostate epithelium is controlled by a cis-element, PSMA enhancer (PSME). PSME contains multiple binding sites for Sox proteins, and in this study, we identified Sox7 protein as a negative regulator of PSMA expression through its interaction with PSME. METHODS: The statistical correlation between Sox7 and PSMA mRNA expression was evaluated using five prostate cancer studies from cBioportal. In vitro and in vivo interaction between Sox7 and PSME was evaluated by chromatin immunoprecipitation (ChIP), electrophoretic mobility shift assay (EMSA), and luciferase reporter assay. Synthetic oligonucleotides were generated to define the sites in PSME that interact with Sox7 protein. Sox7 mutants were generated to identify the region of this protein required to regulate PSMA expression. Sox7 was also stably expressed in LNCaP/C4-2 and 22Rv1 cells to validate the regulation of PSMA expression by Sox7 in vivo. RESULTS: Sox7 mRNA expression negatively correlated with PSMA/FOLH1 and PSMAL/FOLH1B mRNA expression in Broad/Cornell, TCGA and MSKCC studies, but not in two studies containing only metastatic prostate tumors. PC-3 cells mostly expressed the 48.5 KDa isoform 2 of Sox7, and the depletion of this isoform did not restore PSMA expression. Ectopic expression of canonical, wild-type Sox7 in C4-2 and 22Rv1 cells suppressed PSMA protein expression. ChIP assay revealed that canonical Sox7 protein preferentially interacts with PSME in vivo, and EMSA identified the SOX box sites #2 and #4 in PSME as required for its interaction. Sox7 was capable of directly binding to PSME and suppressed PSME-mediated transcription. The NLS regions of Sox7, but not its ß-catenin interacting motif, are essential for this suppressing activity. Furthermore, restoration of wild-type Sox7 expression but not Sox7-NLS mutant in Sox7-null prostate cancer cell lines suppressed PSMA expression. CONCLUSIONS: The inactivation of canonical Sox7 is responsible for the upregulated expression of PSMA in non-metastatic prostate cancer.


Subject(s)
Antigens, Surface/genetics , Enhancer Elements, Genetic/physiology , Gene Expression Regulation, Neoplastic/physiology , Glutamate Carboxypeptidase II/genetics , Prostate/metabolism , Prostatic Neoplasms/metabolism , SOXF Transcription Factors/physiology , Amino Acid Sequence , Base Sequence , Binding Sites , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/chemistry , RNA, Messenger/analysis , SOXF Transcription Factors/chemistry , Wnt Signaling Pathway/physiology
2.
Nucleic Acids Res ; 46(21): 11381-11395, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30335167

ABSTRACT

During embryogenesis, vascular development relies on a handful of transcription factors that instruct cell fate in a distinct sub-population of the endothelium (1). The SOXF proteins that comprise SOX7, 17 and 18, are molecular switches modulating arterio-venous and lymphatic endothelial differentiation (2,3). Here, we show that, in the SOX-F family, only SOX18 has the ability to switch between a monomeric and a dimeric form. We characterized the SOX18 dimer in binding assays in vitro, and using a split-GFP reporter assay in a zebrafish model system in vivo. We show that SOX18 dimerization is driven by a novel motif located in the vicinity of the C-terminus of the DNA binding region. Insertion of this motif in a SOX7 monomer forced its assembly into a dimer. Genome-wide analysis of SOX18 binding locations on the chromatin revealed enrichment for a SOX dimer binding motif, correlating with genes with a strong endothelial signature. Using a SOX18 small molecule inhibitor that disrupts dimerization, we revealed that dimerization is important for transcription. Overall, we show that dimerization is a specific feature of SOX18 that enables the recruitment of key endothelial transcription factors, and refines the selectivity of the binding to discrete genomic locations assigned to endothelial specific genes.


Subject(s)
SOXF Transcription Factors/chemistry , Amino Acid Motifs , Animals , Biosensing Techniques , DNA-Binding Proteins/chemistry , Endothelial Cells/metabolism , Endothelium/metabolism , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/chemistry , Humans , Mice , Mutation , Open Reading Frames , Protein Domains , Protein Multimerization , Zebrafish , Zebrafish Proteins/chemistry
3.
Genome Med ; 10(1): 56, 2018 07 20.
Article in English | MEDLINE | ID: mdl-30029678

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) is a rare disease characterized by distinctive changes in pulmonary arterioles that lead to progressive pulmonary arterial pressures, right-sided heart failure, and a high mortality rate. Up to 30% of adult and 75% of pediatric PAH cases are associated with congenital heart disease (PAH-CHD), and the underlying etiology is largely unknown. There are no known major risk genes for PAH-CHD. METHODS: To identify novel genetic causes of PAH-CHD, we performed whole exome sequencing in 256 PAH-CHD patients. We performed a case-control gene-based association test of rare deleterious variants using 7509 gnomAD whole genome sequencing population controls. We then screened a separate cohort of 413 idiopathic and familial PAH patients without CHD for rare deleterious variants in the top association gene. RESULTS: We identified SOX17 as a novel candidate risk gene (p = 5.5e-7). SOX17 is highly constrained and encodes a transcription factor involved in Wnt/ß-catenin and Notch signaling during development. We estimate that rare deleterious variants contribute to approximately 3.2% of PAH-CHD cases. The coding variants identified include likely gene-disrupting (LGD) and deleterious missense, with most of the missense variants occurring in a highly conserved HMG-box protein domain. We further observed an enrichment of rare deleterious variants in putative targets of SOX17, many of which are highly expressed in developing heart and pulmonary vasculature. In the cohort of PAH without CHD, rare deleterious variants of SOX17 were observed in 0.7% of cases. CONCLUSIONS: These data strongly implicate SOX17 as a new risk gene contributing to PAH-CHD as well as idiopathic/familial PAH. Replication in other PAH cohorts and further characterization of the clinical phenotype will be important to confirm the precise role of SOX17 and better estimate the contribution of genes regulated by SOX17.


Subject(s)
Genetic Variation , Heart Defects, Congenital/complications , Heart Defects, Congenital/genetics , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/genetics , SOXF Transcription Factors/genetics , Adolescent , Adult , Amino Acid Sequence , Child , Child, Preschool , Female , Genetic Predisposition to Disease , Humans , Infant , Male , Risk Factors , SOXF Transcription Factors/chemistry
4.
J Genet ; 97(1): 157-172, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29666335

ABSTRACT

Sox7, Sox17 and Sox18 are the members of the Sry-related high-mobility group box family (SoxF) of transcription factors. SoxF factors regulate endothelial cell fate as well as development and differentiation of blood cells and lymphatic vessels. There is very less information about the functions of these genes in fish. We obtained the full-length cDNA sequence of SoxF genes including Sox7, Sox17 and Sox18 in Cyprinus carpio, where Sox7 and Sox18 had two copies. The construction of a phylogenetic tree showed that these genes were homologous to the genes in other species. Chromosome synteny analysis indicated that the gene order of Sox7 and Sox18 was highly conserved in fish. However, immense change in genomic sequences around Sox17 had taken place. Numerous putative transcription factor binding sites were identified in the 5_ flanking regions of SoxF genes which may be involved in the regulation of the nervous system, vascular epidermal differentiation and embryonic development. The expression levels of SoxF genes were highest in gastrula, and was abundantly expressed in the adult brain.We investigated the expression levels of SoxF genes in five specific parts of the brain. The expression levels of Sox7 and Sox18 were highest in the mesencephalon, while the expression level of Sox17 was highest in the epencephalon. In carp, the expression patterns of SoxF genes indicated a potential function of these genes in neurogenesis and in vascular development. These results provide new information for further studies on the potential functions of SoxF genes in carp.


Subject(s)
Carps/genetics , Gene Expression Regulation, Developmental , SOXF Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , Brain/metabolism , Carps/embryology , Chromosomes/genetics , Embryonic Development/genetics , Genome , Organ Specificity/genetics , Phylogeny , Promoter Regions, Genetic/genetics , SOXF Transcription Factors/chemistry , SOXF Transcription Factors/metabolism , Sequence Alignment , Synteny/genetics
5.
MAbs ; 10(4): 596-606, 2018.
Article in English | MEDLINE | ID: mdl-29648920

ABSTRACT

Antibodies are routinely used to study the activity of transcription factors, using various in vitro and in vivo approaches such as electrophoretic mobility shift assay, enzyme-linked immunosorbent assay, genome-wide method analysis coupled with next generation sequencing, or mass spectrometry. More recently, a new application for antibodies has emerged as crystallisation scaffolds for difficult to crystallise proteins, such as transcription factors. Only in a few rare cases, antibodies have been used to modulate the activity of transcription factors, and there is a real gap in our knowledge on how to efficiently design antibodies to interfere with transcription. The molecular function of transcription factors is underpinned by complex networks of protein-protein interaction and in theory, setting aside intra-cellular delivery challenges, developing antibody-based approaches to modulate transcription factor activity appears a viable option. Here, we demonstrate that antibodies or an antibody single-chain variable region fragments are powerful molecular tools to unravel complex protein-DNA and protein-protein binding mechanisms. In this study, we focus on the molecular mode of action of the transcription factor SOX18, a key modulator of endothelial cell fate during development, as well as an attractive target in certain pathophysiological conditions such as solid cancer metastasis. The engineered antibody we designed inhibits SOX18 transcriptional activity, by interfering specifically with an 8-amino-acid motif in the C-terminal region directly adjacent to α-Helix 3 of SOX18 HMG domain, thereby disrupting protein-protein interaction. This new approach establishes a framework to guide the study of transcription factors interactomes using antibodies as molecular handles.


Subject(s)
SOXF Transcription Factors/analysis , SOXF Transcription Factors/chemistry , Single-Chain Antibodies , Humans
6.
Nat Commun ; 9(1): 1416, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29650961

ABSTRACT

Pulmonary arterial hypertension (PAH) is a rare disorder with a poor prognosis. Deleterious variation within components of the transforming growth factor-ß pathway, particularly the bone morphogenetic protein type 2 receptor (BMPR2), underlies most heritable forms of PAH. To identify the missing heritability we perform whole-genome sequencing in 1038 PAH index cases and 6385 PAH-negative control subjects. Case-control analyses reveal significant overrepresentation of rare variants in ATP13A3, AQP1 and SOX17, and provide independent validation of a critical role for GDF2 in PAH. We demonstrate familial segregation of mutations in SOX17 and AQP1 with PAH. Mutations in GDF2, encoding a BMPR2 ligand, lead to reduced secretion from transfected cells. In addition, we identify pathogenic mutations in the majority of previously reported PAH genes, and provide evidence for further putative genes. Taken together these findings contribute new insights into the molecular basis of PAH and indicate unexplored pathways for therapeutic intervention.


Subject(s)
Adenosine Triphosphatases/chemistry , Aquaporin 1/chemistry , Familial Primary Pulmonary Hypertension/genetics , Growth Differentiation Factors/chemistry , Membrane Transport Proteins/chemistry , Mutation , SOXF Transcription Factors/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adult , Aquaporin 1/genetics , Aquaporin 1/metabolism , Base Sequence , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Case-Control Studies , Familial Primary Pulmonary Hypertension/diagnosis , Familial Primary Pulmonary Hypertension/metabolism , Familial Primary Pulmonary Hypertension/pathology , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Growth Differentiation Factor 2 , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , HEK293 Cells , Humans , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Molecular , Prognosis , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Whole Genome Sequencing
7.
Gene Expr Patterns ; 25-26: 102-108, 2017 11.
Article in English | MEDLINE | ID: mdl-28634012

ABSTRACT

SOX17, a member of SRY-related high-mobility-group box (SOX) family, involves in endoderm formation, angiogenesis and carcinogenesis, and its expression characteristics are different in spermatogenesis among several vertebrates. In this study, we cloned a full-length cDNA sequence of sox17 from Zhikong scallop Chlamys farreri, and determined its expression characteristics in gonad at mRNA and protein levels. The cDNA sequence was 2802 bp in length, predicted to encode a protein of 511 amino acids and contained a conservative HMG-box of SOX family, while lacked the C-terminal region of SOX17 comparing to vertebrates. Semi-quantitative RT-PCR showed that C. farreri sox17 (Cf-sox17) mRNA exhibited a different tissue distribution, and the transcript abundance was the highest in the gonads. In situ hybridization determined that the Cf-sox17 mRNA was located in various germ cells in testis and ovary. Similar result of Cf-SOX17 protein was also observed by immunohistochemical detection. The location in gonad is different from that of mammals and fish in which SOX17 is only located in some specific germ cells. Our finding revealed a different characteristic of sox17 expression in gametogenesis between scallop and vertebrates, which implied that Cf-sox17 may involve in gametogenesis of bivalves and the function may differ from that in vertebrates.


Subject(s)
Gametogenesis , Pectinidae/physiology , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Vertebrates/physiology , Animals , Cloning, Molecular , Female , Gene Expression Regulation, Developmental , Gonads/growth & development , Gonads/metabolism , In Situ Hybridization , Male , Phylogeny , Protein Domains , SOXF Transcription Factors/chemistry , Tissue Distribution
8.
Gene ; 610: 118-126, 2017 Apr 30.
Article in English | MEDLINE | ID: mdl-28132771

ABSTRACT

Essential human proteins; SOX17-HMG domain and beta-catenin uphold a major responsibility for vertebrate gastrulation and embryonic development. Earlier experimental assays document their interaction and states that upon M76A and G103R mutation, their interaction varied. Till date, there was no computational analysis for either of proteins as well as their respective residues for the interaction. The present study extracted and analyzed the experimentally validated 3D models of SOX17-HMG domain and beta-catenin. After analysis of the evolutionarily conserved residues and the sequence-level alteration, the mutated SOX17-HMG protein was re-modeled, demonstrated and energy minimized. Molecular dynamics simulation was performed upon the docked complex of beta-catenin with wild-type and mutant-type protein, individually. Comparable analysis for interaction studies revealed reduction of predominant ionic interactions from 16 (wild-type) to 5 (mutant-type). Glu residues from wild-type protein played a pivotal role forming 50% of the ionic interactions alone. Fascinatingly, statistically significant deductions for several stability calculations deduced the mutant-type protein/complex to form unsteady interaction with beta-catenin. Again, helix-to-coil transition in mutant-type protein supported its weaker conformation. This probe depicts the paramount molecular-level detailed scrutiny for the essential human proteins and disclosure of the mutational analysis, which might tend to hinder the signal transduction. It instigates the future development for the pharmaceutical research.


Subject(s)
Endoderm/embryology , Gastrulation , Molecular Dynamics Simulation , SOXF Transcription Factors/metabolism , beta Catenin/metabolism , HMG-Box Domains , Humans , Mutation , SOXF Transcription Factors/chemistry , SOXF Transcription Factors/genetics , Solvents/chemistry , beta Catenin/chemistry
9.
Semin Cell Dev Biol ; 63: 50-57, 2017 03.
Article in English | MEDLINE | ID: mdl-27470491

ABSTRACT

Cardiovascular development during embryogenesis involves complex changes in gene regulatory networks regulated by a variety of transcription factors. In this review we discuss the various reported roles of the SOXF factors: SOX7, SOX17 and SOX18 in cardiac, vascular and lymphatic development. SOXF factors have pleiotropic roles during these processes, and there is significant redundancy and functional compensation between SOXF family members. Despite this, evidence suggests that there is some specificity in the transcriptional programs they regulate which is necessary to control the differentiation and behaviour of endothelial subpopulations. Furthermore, SOXF factors appear to have an indirect role in regulating cardiac mesoderm specification and differentiation. Understanding how SOXF factors are regulated, as well as their downstream transcriptional target genes, will be important for unravelling their roles in cardiovascular development and related diseases.


Subject(s)
Cardiovascular System/embryology , Cardiovascular System/metabolism , SOXF Transcription Factors/metabolism , Amino Acid Sequence , Animals , Hemangioblasts/metabolism , Humans , Lymphatic Vessels/embryology , Lymphatic Vessels/metabolism , Organogenesis/genetics , SOXF Transcription Factors/chemistry
10.
Nucleic Acids Res ; 44(8): 3922-35, 2016 05 05.
Article in English | MEDLINE | ID: mdl-26939885

ABSTRACT

The transcription factor (TF) SOX18 drives lymphatic vessel development in both embryogenesis and tumour-induced neo-lymphangiogenesis. Genetic disruption of Sox18 in a mouse model protects from tumour metastasis and established the SOX18 protein as a molecular target. Here, we report the crystal structure of the SOX18 DNA binding high-mobility group (HMG) box bound to a DNA element regulating Prox1 transcription. The crystals diffracted to 1.75Å presenting the highest resolution structure of a SOX/DNA complex presently available revealing water structure, structural adjustments at the DNA contact interface and non-canonical conformations of the DNA backbone. To explore alternatives to challenging small molecule approaches for targeting the DNA-binding activity of SOX18, we designed a set of five decoys based on modified Prox1-DNA. Four decoys potently inhibited DNA binding of SOX18 in vitro and did not interact with non-SOX TFs. Serum stability, nuclease resistance and thermal denaturation assays demonstrated that a decoy circularized with a hexaethylene glycol linker and terminal phosphorothioate modifications is most stable. This SOX decoy also interfered with the expression of a luciferase reporter under control of a SOX18-dependent VCAM1 promoter in COS7 cells. Collectively, we propose SOX decoys as potential strategy for inhibiting SOX18 activity to disrupt tumour-induced neo-lymphangiogenesis.


Subject(s)
DNA/chemistry , Homeodomain Proteins/genetics , SOXF Transcription Factors/antagonists & inhibitors , SOXF Transcription Factors/chemistry , Tumor Suppressor Proteins/genetics , Animals , COS Cells , Chlorocebus aethiops , DNA/metabolism , Gene Expression Regulation , Mice , Nucleic Acid Conformation , Oligonucleotides , SOX Transcription Factors/chemistry , SOX Transcription Factors/metabolism , SOXF Transcription Factors/metabolism , Transcription, Genetic
11.
Structure ; 22(9): 1274-1286, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25126959

ABSTRACT

In pluripotent cells, OCT4 associates with SOX2 to maintain pluripotency or with SOX17 to induce primitive endoderm commitment. The OCT4-SOX2 and OCT4-SOX17 combinations bind mutually exclusive to two distinct composite DNA elements, known as the "canonical" and "compressed" motifs, respectively. The structural basis for the OCT4-SOX17 cooperativity is unknown. Whereas SOX17 has been engineered to replace SOX2 in the pluripotency circuitry, all generated SOX2 mutants have failed to act like SOX17. From molecular simulations, we revealed the OCT4-SOX17 interaction interface and elucidated the SOX-dependent motif preference of OCT4. Moreover, we designed a SOX2 mutant that we predicted and confirmed experimentally to bind cooperatively with OCT4 to the compressed motif. Ultimately, we found a strong correlation between the experimental and calculated relative cooperative-binding free energies of 12 OCT4-SOX-DNA complexes. Therefore, we validated the OCT4-SOX interfaces and demonstrated that in silico design of DNA-binding cooperativity is suitable for altering transcriptional circuitries.


Subject(s)
HMGB Proteins/chemistry , Octamer Transcription Factor-3/chemistry , SOXB1 Transcription Factors/chemistry , SOXF Transcription Factors/chemistry , Stem Cells/physiology , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation , Consensus Sequence , DNA/chemistry , HMGB Proteins/genetics , Hydrophobic and Hydrophilic Interactions , Mice , Molecular Dynamics Simulation , Molecular Sequence Data , Octamer Transcription Factor-3/genetics , Protein Binding , Protein Interaction Domains and Motifs , SOXB1 Transcription Factors/genetics , SOXF Transcription Factors/genetics , Thermodynamics
12.
Protein Pept Lett ; 20(4): 481-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23061670

ABSTRACT

Sry-related box (Sox) transcription factors share a conserved high-mobility-group box domain (HMG-domain) that binds DNA in the minor groove and bends DNA for further assembly of transcriptional machineries. During organogenesis, each member of the Sox family triggers a specific cell lineage differentiation, indicating that their interactions with DNA are different from each other. Therefore, investigating structural rearrangement of each Sox transcription factor HMG-domain upon binding to DNA would help to elucidate the distinctive molecular mechanism by which they interact with DNA. Previous studies have determined the crystal structures of Sox2 HMG-domain/DNA, Sox4 HMGdomain/ DNA, Sox9 HMG-domain/DNA and Sox17 HMG-domain/DNA complexes. However, major gaps remain in the structural information on the Sox transcription factor HMG-domains. Here, we report the crystal structure of the human Sox17 HMG-domain alone at 2.4 A resolution. Comparing this structure and the structure of the mouse Sox17 HMGdomain/ DNA complex provides structural understanding of the mechanism of Sox17 binding to DNA. Specifically, after electrostatic interactions attract Sox17 to DNA, Asn73, Ser99, and Trp106 form hydrogen bonds with DNA, Arg70, Lys80, Arg83, His94, and Asn95 on Sox17 undergo conformational changes and form hydrogen bonds with DNA, contributing to the electrostatic interaction between Sox17 and DNA.


Subject(s)
DNA/metabolism , SOXF Transcription Factors/chemistry , SOXF Transcription Factors/metabolism , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , HMG-Box Domains , Humans , Hydrogen Bonding , Mice , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Static Electricity
13.
Biochemistry (Mosc) ; 78(11): 1287-92, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24460943

ABSTRACT

SOX18 transcription factor plays important roles in a range of biological processes such as vasculogenesis, hair follicle development, lymphangiogenesis, atherosclerosis, and angiogenesis. In this paper we present the generation of a novel SOX18 dominant-negative mutant (SOX18DN) encoding truncated SOX18 protein that lacks a trans-activation domain. We show that both wild-type SOX18 (SOX18wt) and truncated human SOX18 proteins are able to bind to their consensus sequence in vitro. Functional analysis confirmed that SOX18wt has potent trans-activation properties, while SOX18DN displays dominant-negative effect. We believe that these SOX18wt and SOX18DN expression constructs could be successfully used for further characterization of the function of this protein.


Subject(s)
Mutation , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Base Sequence , Genes, Reporter , HeLa Cells , Humans , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , SOXF Transcription Factors/chemistry , Transcriptional Activation
14.
Biochem J ; 443(1): 39-47, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22181698

ABSTRACT

It has recently been proposed that the sequence preferences of DNA-binding TFs (transcription factors) can be well described by models that include the positional interdependence of the nucleotides of the target sites. Such binding models allow for multiple motifs to be invoked, such as principal and secondary motifs differing at two or more nucleotide positions. However, the structural mechanisms underlying the accommodation of such variant motifs by TFs remain elusive. In the present study we examine the crystal structure of the HMG (high-mobility group) domain of Sox4 [Sry (sex-determining region on the Y chromosome)-related HMG box 4] bound to DNA. By comparing this structure with previously solved structures of Sox17 and Sox2, we observed subtle conformational differences at the DNA-binding interface. Furthermore, using quantitative electrophoretic mobility-shift assays we validated the positional interdependence of two nucleotides and the presence of a secondary Sox motif in the affinity landscape of Sox4. These results suggest that a concerted rearrangement of two interface amino acids enables Sox4 to accommodate primary and secondary motifs. The structural adaptations lead to altered dinucleotide preferences that mutually reinforce each other. These analyses underline the complexity of the DNA recognition by TFs and provide an experimental validation for the conceptual framework of positional interdependence and secondary binding motifs.


Subject(s)
DNA/chemistry , Macromolecular Substances/chemistry , SOXC Transcription Factors/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic , HMGB Proteins/chemistry , Laminin/genetics , Mice , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , SOXF Transcription Factors/chemistry
15.
J Mol Biol ; 388(3): 619-30, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19328208

ABSTRACT

Sox17 regulates endodermal lineage commitment and is thought to function antagonistically to the pluripotency determinant Sox2. To investigate the biochemical basis for the distinct functions of Sox2 and Sox17, we solved the crystal structure of the high mobility group domain of Sox17 bound to a DNA element derived from the Lama1 enhancer using crystals diffracting to 2.7 A resolution. Sox17 targets the minor groove and bends the DNA by approximately 80 degrees . The DNA architecture closely resembles the one seen for Sox2/DNA structures, suggesting that the degree of bending is conserved between both proteins and nucleotide substitutions have only marginal effects on the bending topology. Accordingly, affinities of Sox2 and Sox17 for the Lama1 element were found to be identical. However, when the Oct1 contact interface of Sox2 is compared with the corresponding region of Sox17, a significantly altered charge distribution is observed, suggesting differential co-factor recruitment that may explain their biological distinctiveness.


Subject(s)
HMGB Proteins/chemistry , SOXF Transcription Factors/chemistry , Amino Acid Sequence , Circular Dichroism , Crystallography, X-Ray , DNA/metabolism , Electrophoretic Mobility Shift Assay , Laminin/genetics , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , Protein Structure, Tertiary , Sequence Alignment
16.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 64(Pt 12): 1184-7, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19052383

ABSTRACT

Sox17 is a member of the SRY-related high-mobility group (HMG) of transcription factors that have been shown to direct endodermal differentiation in early mammalian development. The LAMA1 gene encoding the alpha-chain of laminin-1 has been reported to be directly bound and regulated by Sox17. This paper describes the details of initial crystallization attempts with the HMG domain of mouse Sox17 (mSox17-HMG) with a 16-mer DNA element derived from the LAMA1 enhancer and optimization strategies to obtain a better diffracting crystal. The best diffracting crystal was obtained in a condition containing 0.1 M Tris-HCl pH 7.4, 0.2 M MgCl(2), 30% PEG 3350 using the hanging-drop vapour-diffusion method. A highly redundant in-house data set was collected to 2.75 A resolution with 99% completeness. The presence of the mSox17-HMG-DNA complex within the crystals was confirmed and Matthews analysis indicated the presence of one complex per asymmetric unit.


Subject(s)
DNA/chemistry , HMGB Proteins/chemistry , SOXF Transcription Factors/chemistry , Animals , Base Sequence , Cloning, Molecular , Crystallization , Crystallography, X-Ray , DNA/metabolism , HMGB Proteins/isolation & purification , HMGB Proteins/metabolism , Mice , Molecular Sequence Data , Protein Structure, Tertiary , SOXF Transcription Factors/isolation & purification , SOXF Transcription Factors/metabolism , X-Ray Diffraction
SELECTION OF CITATIONS
SEARCH DETAIL
...