Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Endocrinol (Lausanne) ; 13: 1029177, 2022.
Article in English | MEDLINE | ID: mdl-36568077

ABSTRACT

Y-encoded transcription factor SRY initiates male differentiation in therian mammals. This factor contains a high-mobility-group (HMG) box, which mediates sequence-specific DNA binding with sharp DNA bending. A companion article in this issue described sex-reversal mutations at box position 72 (residue 127 in human SRY), invariant as Tyr among mammalian orthologs. Although not contacting DNA, the aromatic ring seals the domain's minor wing at a solvent-exposed junction with a basic tail. A seeming paradox was posed by the native-like biochemical properties of inherited Swyer variant Y72F: its near-native gene-regulatory activity is consistent with the father's male development, but at odds with the daughter's XY female somatic phenotype. Surprisingly, aromatic rings (Y72, F72 or W72) confer higher transcriptional activity than do basic or polar side chains generally observed at solvated DNA interfaces (Arg, Lys, His or Gln). Whereas biophysical studies (time-resolved fluorescence resonance energy transfer and heteronuclear NMR spectroscopy) uncovered only subtle perturbations, dissociation of the Y72F complex was markedly accelerated relative to wild-type. Studies of protein-DNA solvation by molecular-dynamics (MD) simulations of an homologous high-resolution crystal structure (SOX18) suggest that Y72 para-OH anchors a network of water molecules at the tail-DNA interface, perturbed in the variant in association with nonlocal conformational fluctuations. Loss of the Y72 anchor among SRY variants presumably "unclamps" its basic tail, leading to (a) rapid DNA dissociation despite native affinity and (b) attenuated transcriptional activity at the edge of sexual ambiguity. Conservation of Y72 suggests that this water-mediated clamp operates generally among SRY and metazoan SOX domains.


Subject(s)
Sex Determination Processes , Transcription Factors , Animals , Female , Humans , Male , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation , Mammals/genetics , Mammals/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Sex Determination Processes/genetics , Sex Determination Processes/physiology
2.
Front Endocrinol (Lausanne) ; 13: 945030, 2022.
Article in English | MEDLINE | ID: mdl-35957822

ABSTRACT

Male sex determination in mammals is initiated by SRY, a Y-encoded transcription factor. The protein contains a high-mobility-group (HMG) box mediating sequence-specific DNA bending. Mutations causing XY gonadal dysgenesis (Swyer syndrome) cluster in the box and ordinarily arise de novo. Rare inherited variants lead to male development in one genetic background (the father) but not another (his sterile XY daughter). De novo and inherited mutations occur at an invariant Tyr adjoining the motif's basic tail (box position 72; Y127 in SRY). In SRY-responsive cell lines CH34 and LNCaP, de novo mutations Y127H and Y127C reduced SRY activity (as assessed by transcriptional activation of principal target gene Sox9) by 5- and 8-fold, respectively. Whereas Y127H impaired testis-specific enhancer assembly, Y127C caused accelerated proteasomal proteolysis; activity was in part rescued by proteasome inhibition. Inherited variant Y127F was better tolerated: its expression was unperturbed, and activity was reduced by only twofold, a threshold similar to other inherited variants. Biochemical studies of wild-type (WT) and variant HMG boxes demonstrated similar specific DNA affinities (within a twofold range), with only subtle differences in sharp DNA bending as probed by permutation gel electrophoresis and fluorescence resonance-energy transfer (FRET); thermodynamic stabilities of the free boxes were essentially identical. Such modest perturbations are within the range of species variation. Whereas our cell-based findings rationalize the de novo genotype-phenotype relationships, a molecular understanding of inherited mutation Y127F remains elusive. Our companion study uncovers cryptic biophysical perturbations suggesting that the para-OH group of Y127 anchors a novel water-mediated DNA clamp.


Subject(s)
Gonadal Dysgenesis, 46,XY , Animals , Cell Line , DNA/metabolism , Fluorescence Resonance Energy Transfer , Gonadal Dysgenesis, 46,XY/genetics , Humans , Male , Mammals/genetics , Sex-Determining Region Y Protein/chemistry , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism
3.
J Biol Chem ; 291(42): 22173-22195, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27576690

ABSTRACT

A general problem is posed by analysis of transcriptional thresholds governing cell fate decisions in metazoan development. A model is provided by testis determination in therian mammals. Its key step, Sertoli cell differentiation in the embryonic gonadal ridge, is initiated by SRY, a Y-encoded architectural transcription factor. Mutations in human SRY cause gonadal dysgenesis leading to XY female development (Swyer syndrome). Here, we have characterized an inherited mutation compatible with either male or female somatic phenotypes as observed in an XY father and XY daughter, respectively. The mutation (a crevice-forming substitution at a conserved back surface of the SRY high mobility group box) markedly destabilizes the domain but preserves specific DNA affinity and induced DNA bend angle. On transient transfection of diverse human and rodent cell lines, the variant SRY exhibited accelerated proteasomal degradation (relative to wild type) associated with increased ubiquitination; in vitro susceptibility to ubiquitin-independent ("default") cleavage by the 20S core proteasome was unchanged. The variant's gene regulatory activity (as assessed in a cellular model of the rat embryonic XY gonadal ridge) was reduced by 2-fold relative to wild-type SRY at similar levels of mRNA expression. Chemical proteasome inhibition restored native-like SRY expression and transcriptional activity in association with restored occupancy of a sex-specific enhancer element in principal downstream gene Sox9, demonstrating that the variant SRY exhibits essentially native activity on a per molecule basis. Our findings define a novel mechanism of impaired organogenesis, accelerated ubiquitin-directed proteasomal degradation of a master transcription factor leading to a developmental decision poised at the edge of ambiguity.


Subject(s)
Disorder of Sex Development, 46,XY/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Sertoli Cells/metabolism , Sex-Determining Region Y Protein/metabolism , Ubiquitination , Animals , Disorder of Sex Development, 46,XY/genetics , Female , Humans , Male , Proteasome Endopeptidase Complex/genetics , Rats , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sex-Determining Region Y Protein/genetics
4.
J Biol Chem ; 289(47): 32410-29, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25258310

ABSTRACT

Human testis determination is initiated by SRY, a Y-encoded architectural transcription factor. Mutations in SRY cause 46 XY gonadal dysgenesis with female somatic phenotype (Swyer syndrome) and confer a high risk of malignancy (gonadoblastoma). Such mutations cluster in the SRY high mobility group (HMG) box, a conserved motif of specific DNA binding and bending. To explore structure-function relationships, we constructed all possible substitutions at a site of clinical mutation (W70L). Our studies thus focused on a core aromatic residue (position 15 of the consensus HMG box) that is invariant among SRY-related HMG box transcription factors (the SOX family) and conserved as aromatic (Phe or Tyr) among other sequence-specific boxes. In a yeast one-hybrid system sensitive to specific SRY-DNA binding, the variant domains exhibited reduced (Phe and Tyr) or absent activity (the remaining 17 substitutions). Representative nonpolar variants with partial or absent activity (Tyr, Phe, Leu, and Ala in order of decreasing side-chain volume) were chosen for study in vitro and in mammalian cell culture. The clinical mutation (Leu) was found to markedly impair multiple biochemical and cellular activities as respectively probed through the following: (i) in vitro assays of specific DNA binding and protein stability, and (ii) cell culture-based assays of proteosomal degradation, nuclear import, enhancer DNA occupancy, and SRY-dependent transcriptional activation. Surprisingly, however, DNA bending is robust to this or the related Ala substitution that profoundly impairs box stability. Together, our findings demonstrate that the folding, trafficking, and gene-regulatory function of SRY requires an invariant aromatic "buttress" beneath its specific DNA-bending surface.


Subject(s)
DNA-Binding Proteins/chemistry , DNA/chemistry , Nucleic Acid Conformation , Protein Structure, Tertiary , Sex-Determining Region Y Protein/chemistry , Amino Acid Sequence , Amino Acids, Aromatic/chemistry , Amino Acids, Aromatic/genetics , Amino Acids, Aromatic/metabolism , Animals , Binding Sites/genetics , Blotting, Western , Cell Line , Circular Dichroism , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescence Resonance Energy Transfer , Humans , Male , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding/genetics , Protein Folding , Sequence Homology, Amino Acid , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Structure-Activity Relationship , Thermodynamics , Transcriptional Activation
5.
Proc Natl Acad Sci U S A ; 110(38): E3567-76, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-24003159

ABSTRACT

Human testis determination is initiated by SRY (sex determining region on Y chromosome). Mutations in SRY cause gonadal dysgenesis with female somatic phenotype. Two subtle variants (V60L and I90M in the high-mobility group box) define inherited alleles shared by an XY sterile daughter and fertile father. Whereas specific DNA binding and bending are unaffected in a rat embryonic pre-Sertoli cell line, the variants exhibited selective defects in nucleocytoplasmic shuttling due to impaired nuclear import (V60L; mediated by Exportin-4) or export (I90M; mediated by chromosome region maintenance 1). Decreased shuttling limits nuclear accumulation of phosphorylated (activated) SRY, in turn reducing occupancy of DNA sites regulating Sertoli-cell differentiation [the testis-specific SRY-box 9 (Sox9) enhancer]. Despite distinct patterns of biochemical and cell-biological perturbations, V60L and I90M each attenuated Sox9 expression in transient transfection assays by twofold. Such attenuation was also observed in studies of V60A, a clinical variant associated with ovotestes and hence ambiguity between divergent cell fates. This shared twofold threshold is reminiscent of autosomal syndromes of transcription-factor haploinsufficiency, including XY sex reversal associated with mutations in SOX9. Our results demonstrate that nucleocytoplasmic shuttling of SRY is necessary for robust initiation of testicular development. Although also characteristic of ungulate orthologs, such shuttling is not conserved among rodents wherein impaired nuclear export of the high-mobility group box and import-dependent phosphorylation are compensated by a microsatellite-associated transcriptional activation domain. Human sex reversal due to subtle defects in the nucleocytoplasmic shuttling of SRY suggests that its transcriptional activity lies near the edge of developmental ambiguity.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation, Developmental/genetics , Gonadal Dysgenesis, 46,XY/genetics , SOX9 Transcription Factor/metabolism , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Testis/growth & development , Active Transport, Cell Nucleus/physiology , Blotting, Western , Cell Differentiation/physiology , Cell Line , Chromatin Immunoprecipitation , Female , Humans , Immunohistochemistry , Male , Models, Molecular , Protein Conformation , SOX9 Transcription Factor/genetics , Sertoli Cells/cytology
6.
Proc Natl Acad Sci U S A ; 110(33): E3061-70, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23901118

ABSTRACT

The male program of therian mammals is determined by Sry, a transcription factor encoded by the Y chromosome. Specific DNA binding is mediated by a high mobility group (HMG) box. Expression of Sry in the gonadal ridge activates a Sox9-dependent gene regulatory network leading to testis formation. A subset of Sry alleles in superfamily Muroidea (order Rodentia) is remarkable for insertion of an unstable DNA microsatellite, most commonly encoding (as in mice) a CAG repeat-associated glutamine-rich domain. We provide evidence, based on an embryonic pre-Sertoli cell line, that this domain functions at a threshold length as a genetic capacitor to facilitate accumulation of variation elsewhere in the protein, including the HMG box. The glutamine-rich domain compensates for otherwise deleterious substitutions in the box and absence of nonbox phosphorylation sites to ensure occupancy of DNA target sites. Such compensation enables activation of a male transcriptional program despite perturbations to the box. Whereas human SRY requires nucleocytoplasmic shuttling and coupled phosphorylation, mouse Sry contains a defective nuclear export signal analogous to a variant human SRY associated with inherited sex reversal. We propose that the rodent glutamine-rich domain has (i) fostered accumulation of cryptic intragenic variation and (ii) enabled unmasking of such variation due to DNA replicative slippage. This model highlights genomic contingency as a source of protein novelty at the edge of developmental ambiguity and may underlie emergence of non-Sry-dependent sex determination in the radiation of Muroidea.


Subject(s)
Biological Evolution , DNA/metabolism , Gene Expression Regulation/physiology , Gene Regulatory Networks/genetics , Rodentia/genetics , Sex Determination Processes/genetics , Sex-Determining Region Y Protein/genetics , Animals , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Circular Dichroism , DNA/genetics , Fluorescence Resonance Energy Transfer , Gene Expression Regulation/genetics , HMG-Box Domains/genetics , Humans , Immunohistochemistry , Male , Mice , Microsatellite Repeats/genetics , Protein Structure, Tertiary/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sex-Determining Region Y Protein/chemistry , Sex-Determining Region Y Protein/metabolism , Spectrometry, Fluorescence , Trinucleotide Repeats/genetics
7.
J Biol Chem ; 286(42): 36787-807, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21849498

ABSTRACT

Mammalian testis-determining factor SRY contains a high mobility group box, a conserved eukaryotic motif of DNA bending. Mutations in SRY cause XY gonadal dysgenesis and somatic sex reversal. Although such mutations usually arise de novo in spermatogenesis, some are inherited and so specify male development in one genetic background (the father) but not another (the daughter). Here, we describe the biophysical properties of a representative inherited mutation, V60L, within the minor wing of the L-shaped domain (box position 5). Although the stability and DNA binding properties of the mutant domain are similar to those of wild type, studies of SRY-induced DNA bending by subnanosecond time-resolved fluorescence resonance energy transfer (FRET) revealed enhanced conformational fluctuations leading to long range variation in bend angle. (1)H NMR studies of the variant protein-DNA complex demonstrated only local perturbations near the mutation site. Because the minor wing of SRY folds on DNA binding, the inherited mutation presumably hinders induced fit. Stopped-flow FRET studies indicated that such frustrated packing leads to accelerated dissociation of the bent complex. Studies of SRY-directed transcriptional regulation in an embryonic gonadal cell line demonstrated partial activation of downstream target Sox9. Our results have demonstrated a nonlocal coupling between DNA-directed protein folding and protein-directed DNA bending. Perturbation of this coupling is associated with a genetic switch poised at the threshold of activity.


Subject(s)
Amino Acid Substitution , DNA/chemistry , Gonadal Dysgenesis, 46,XY , Mutation, Missense , Nucleic Acid Conformation , Protein Folding , Sex-Determining Region Y Protein/chemistry , Animals , Cell Line , DNA/metabolism , Humans , Magnetic Resonance Spectroscopy , Male , Protein Structure, Tertiary , Rodentia , SOX9 Transcription Factor/chemistry , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Structure-Activity Relationship , Transcription, Genetic/genetics
8.
Proc Natl Acad Sci U S A ; 103(11): 4022-7, 2006 Mar 14.
Article in English | MEDLINE | ID: mdl-16537479

ABSTRACT

Ribonucleotide reductase catalyzes a crucial step in de novo DNA synthesis and is allosterically controlled by relative levels of dNTPs to maintain a balanced pool of deoxynucleoside triphosphates in the cell. In eukaryotes, the enzyme comprises a heterooligomer of alpha(2) and beta(2) subunits. The alpha subunit, Rnr1, contains catalytic and regulatory sites. Here, we report the only x-ray structures of the eukaryotic alpha subunit of ribonucleotide reductase from Saccharomyces cerevisiae. The structures of the apo-, AMPPNP only-, AMPPNP-CDP-, AMPPNP-UDP-, dGTP-ADP- and TTP-GDP-bound complexes give insight into substrate and effector binding and specificity cross-talk. These are Class I structures with the only fully ordered catalytic sites, including loop 2, a stretch of polypeptide that spans specificity and catalytic sites, conferring specificity. Binding of specificity effector rearranges loop 2; in our structures, this rearrangement moves P294, a residue unique to eukaryotes, out of the catalytic site, accommodating substrate binding. Substrate binding further rearranges loop 2. Cross-talk, by which effector binding regulates substrate preference, occurs largely through R293 and Q288 of loop 2, which are analogous to residues in Thermotoga maritima that mediate cross-talk. However loop-2 conformations and residue-substrate interactions differ substantially between yeast and T. maritima. In most effector-substrate complexes, water molecules help mediate substrate-loop 2 interactions. Finally, the substrate ribose binds with its 3' hydroxyl closer than its 2' hydroxyl to C218 of the catalytic redox pair. We also see a conserved water molecule at the catalytic site in all our structures, near the ribose 2' hydroxyl.


Subject(s)
Deoxyribonucleotides/metabolism , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Catalytic Domain , Crystallography, X-Ray , Models, Molecular , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Substrate Specificity , Thermotoga maritima/enzymology
9.
Proc Natl Acad Sci U S A ; 103(11): 4028-33, 2006 Mar 14.
Article in English | MEDLINE | ID: mdl-16537480

ABSTRACT

Ribonucleotide reductase (RNR) catalyzes the conversion of nucleoside diphosphates to deoxynucleoside diphosphates. Crucial for rapidly dividing cells, RNR is a target for cancer therapy. In eukaryotes, RNR comprises a heterooligomer of alpha(2) and beta(2) subunits. Rnr1, the alpha subunit, contains regulatory and catalytic sites; Rnr2, the beta subunit (in yeast, a heterodimer of Rnr2 and Rnr4), houses the diferric-tyrosyl radical crucial for catalysis. Here, we present three x-ray structures of eukaryotic Rnr1 from Saccharomyces cerevisiae: one bound to gemcitabine diphosphate (GemdP), the active metabolite of the mechanism-based chemotherapeutic agent gemcitabine; one with an Rnr2-derived peptide, and one with an Rnr4-derived peptide. Our structures reveal that GemdP binds differently from its analogue, cytidine diphosphate; because of unusual interactions of the geminal fluorines, the ribose and base of GemdP shift substantially, and loop 2, which mediates substrate specificity, adopts different conformations when binding to GemdP and cytidine diphosphate. The Rnr2 and Rnr4 peptides, which block RNR assembly, bind differently from each other but have unique modes of binding not seen in prokaryotic RNR. The Rnr2 peptide adopts a conformation similar to that previously reported from an NMR study for a mouse Rnr2-based peptide. In yeast, the Rnr2 peptide binds at subsites consisting of residues that are highly conserved among yeast, mouse, and human Rnr1s, suggesting that the mode of Rnr1-Rnr2 binding is conserved among eukaryotes. These structures provide new insights into subunit assembly and a framework for structure-based drug design targeting RNR.


Subject(s)
Deoxycytidine/analogs & derivatives , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray , Deoxycytidine/metabolism , Deoxycytosine Nucleotides/metabolism , Dimerization , Escherichia coli/enzymology , Escherichia coli/genetics , Humans , Mice , Models, Molecular , Molecular Sequence Data , Protein Structure, Quaternary , Protein Subunits , Ribonucleotide Reductases/genetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid , Static Electricity , Substrate Specificity , Gemcitabine
10.
Biochemistry ; 43(26): 8568-78, 2004 Jul 06.
Article in English | MEDLINE | ID: mdl-15222768

ABSTRACT

Sml1p is a small 104-amino acid protein from Saccharomyces cerevisiae that binds to the large subunit (Rnr1p) of the ribonucleotide reductase complex (RNR) and inhibits its activity. During DNA damage, S phase, or both, RNR activity must be tightly regulated, since failure to control the cellular level of dNTP pools may lead to genetic abnormalities, such as genome rearrangements, or even cell death. Structural characterization of Sml1p is an important step in understanding the regulation of RNR. Until now the oligomeric state of Sml1p was unknown. Mass spectrometric analysis of wild-type Sml1p revealed an intermolecular disulfide bond involving the cysteine residue at position 14 of the primary sequence. To determine whether disulfide bonding is essential for Sml1p oligomerization, we mutated the Cys14 to serine. Sedimentation equilibrium measurements in the analytical ultracentrifuge show that both wild-type and C14S Sml1p exist as dimers in solution, indicating that the dimerization is not a result of a disulfide bond. Further studies of several truncated Sml1p mutants revealed that the N-terminal 8-20 residues are responsible for dimerization. Unfolding/refolding studies of wild-type and C14S Sml1p reveal that both proteins refold reversibly and have almost identical unfolding/refolding profiles. It appears that Sml1p is a two-domain protein where the N-terminus is responsible for dimerization and the C-terminus for binding and inhibiting Rnr1p activity.


Subject(s)
Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae/chemistry , Amino Acid Sequence , Biophysical Phenomena , Biophysics , Circular Dichroism , Cloning, Molecular , Cysteine/chemistry , Dimerization , Disulfides , Dose-Response Relationship, Drug , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Models, Statistical , Molecular Sequence Data , Protein Denaturation , Protein Folding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Spectrometry, Mass, Electrospray Ionization , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL
...