Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Article in English | MEDLINE | ID: mdl-33975946

ABSTRACT

Compaction of bulky DNA is a universal issue for all DNA-based life forms. Chloroplast nucleoids (chloroplast DNA-protein complexes) are critical for chloroplast DNA maintenance and transcription, thereby supporting photosynthesis, but their detailed structure remains enigmatic. Our proteomic analysis of chloroplast nucleoids of the green alga Chlamydomonas reinhardtii identified a protein (HBD1) with a tandem repeat of two DNA-binding high mobility group box (HMG-box) domains, which is structurally similar to major mitochondrial nucleoid proteins transcription factor A, mitochondrial (TFAM), and ARS binding factor 2 protein (Abf2p). Disruption of the HBD1 gene by CRISPR-Cas9-mediated genome editing resulted in the scattering of chloroplast nucleoids. This phenotype was complemented when intact HBD1 was reintroduced, whereas a truncated HBD1 with a single HMG-box domain failed to complement the phenotype. Furthermore, ectopic expression of HBD1 in the mitochondria of yeast Δabf2 mutant successfully complemented the defects, suggesting functional similarity between HBD1 and Abf2p. Furthermore, in vitro assays of HBD1, including the electrophoretic mobility shift assay and DNA origami/atomic force microscopy, showed that HBD1 is capable of introducing U-turns and cross-strand bridges, indicating that proteins with two HMG-box domains would function as DNA clips to compact DNA in both chloroplast and mitochondrial nucleoids.


Subject(s)
Chlamydomonas reinhardtii/genetics , Chloroplast Proteins/genetics , DNA, Chloroplast/genetics , Genome, Chloroplast/genetics , HMG-Box Domains/genetics , Tandem Repeat Sequences/genetics , Chlamydomonas reinhardtii/metabolism , Chloroplast Proteins/classification , Chloroplast Proteins/metabolism , DNA, Chloroplast/metabolism , Gene Expression Regulation , Mass Spectrometry/methods , Mutation , Phylogeny , Protein Binding , Proteomics/methods
2.
Proc Natl Acad Sci U S A ; 118(2)2021 01 12.
Article in English | MEDLINE | ID: mdl-33443157

ABSTRACT

The sex-determining region on the Y chromosome (SRY) is thought to be the central genetic element of male sex development in mammals. Pathogenic modifications within the SRY gene are associated with a male-to-female sex reversal syndrome in humans and other mammalian species, including rabbits and mice. However, the underlying mechanisms are largely unknown. To understand the biological function of the SRY gene, a site-directed mutational analysis is required to investigate associated phenotypic changes at the molecular, cellular, and morphological level. Here, we successfully generated a knockout of the porcine SRY gene by microinjection of two CRISPR-Cas ribonucleoproteins, targeting the centrally located "high mobility group" (HMG), followed by a frameshift mutation of the downstream SRY sequence. This resulted in the development of genetically male (XY) pigs with complete external and internal female genitalia, which, however, were significantly smaller than in 9-mo-old age-matched control females. Quantitative digital PCR analysis revealed a duplication of the SRY locus in Landrace pigs similar to the known palindromic duplication in Duroc breeds. Our study demonstrates the central role of the HMG domain in the SRY gene in male porcine sex determination. This proof-of-principle study could assist in solving the problem of sex preference in agriculture to improve animal welfare. Moreover, it establishes a large animal model that is more comparable to humans with regard to genetics, physiology, and anatomy, which is pivotal for longitudinal studies to unravel mammalian sex determination and relevant for the development of new interventions for human sex development disorders.


Subject(s)
Sex Determination Processes/genetics , Sex-Determining Region Y Protein/genetics , Sex-Determining Region Y Protein/metabolism , Amino Acid Sequence/genetics , Animals , DNA-Binding Proteins/genetics , Disorders of Sex Development/genetics , Frameshift Mutation/genetics , Genes, sry/genetics , HMG-Box Domains/genetics , Male , Mutation/genetics , Nuclear Proteins/genetics , Proof of Concept Study , Protein Domains/genetics , Swine/genetics , Transcription Factors/genetics , Y Chromosome/genetics
3.
Gene Expr Patterns ; 36: 119112, 2020 06.
Article in English | MEDLINE | ID: mdl-32259660

ABSTRACT

Sox protein family is characterized by the presence of the conserved high-mobility group (HMG) box. Sox transcription factors are involved in diverse developmental process in animals, including sex-determination, organogenesis, embryogenesis, neurogenesis, and cell fate decision. In this study, 23 Sox genes were identified based on the Culter alburnus whole-genome sequence and categorized into six subfamilies according to the conserved HMG-box domain. The duplicates of four members revealed that Sox genes in the teleost fishes underwent significant expansion. Moreover, their expression pattern in gonad tissues were analyzed by RNA-seq and qRT-PCR, and Sox9b was determined as a key gene that was essential for testis development. This current study will provide new insight into the role of Sox gene family in fish sex determination and differentiation.


Subject(s)
Cyprinidae/genetics , Cyprinidae/metabolism , HMG-Box Domains/genetics , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , Amino Acid Sequence , Animals , Embryonic Development , Fish Proteins/genetics , Fish Proteins/metabolism , Gene Expression Regulation, Developmental , Genome-Wide Association Study , RNA-Seq , Sex Determination Processes , Transcriptome , Whole Genome Sequencing
4.
FEBS J ; 286(24): 4951-4963, 2019 12.
Article in English | MEDLINE | ID: mdl-31323153

ABSTRACT

Capicua (CIC) is a transcriptional repressor and functions downstream of the receptor tyrosine kinase (RTK) signaling pathway. Somatic mutations found in the HMG-box DNA binding domain in CIC have been implicated in several cancers such as oligodendroglioma, oligoastrocytoma, and adenocarcinoma. However, the molecular basis of the DNA binding of CIC and the effect of the somatic mutations found in cancers on DNA binding have not been investigated. Here, we report the crystal structure of the HMG-box domain of CIC complexed with its target DNA, the promoter of Ets Translocation Variant 5 (ETV5). The structure shows that the HMG-box domain has an L-shaped structure and recognizes the minor groove leading to DNA bending. Our structure combined with an electrophoretic mobility shift assay (EMSA) revealed that cancer-associated mutations in the HMG-box domain abrogate the interaction with DNA. These results provide the molecular insight into the DNA binding of CIC and reveal the effects of carcinogenic mutations on DNA binding.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , HMG-Box Domains/physiology , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Electrophoretic Mobility Shift Assay , HMG-Box Domains/genetics , Humans , Mutation/genetics , Neoplasms/chemistry , Neoplasms/genetics , Neoplasms/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factors/genetics
5.
Am J Hum Genet ; 104(2): 246-259, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30661772

ABSTRACT

SOX4, together with SOX11 and SOX12, forms group C of SRY-related (SOX) transcription factors. They play key roles, often in redundancy, in multiple developmental pathways, including neurogenesis and skeletogenesis. De novo SOX11 heterozygous mutations have been shown to cause intellectual disability, growth deficiency, and dysmorphic features compatible with mild Coffin-Siris syndrome. Using trio-based exome sequencing, we here identify de novo SOX4 heterozygous missense variants in four children who share developmental delay, intellectual disability, and mild facial and digital morphological abnormalities. SOX4 is highly expressed in areas of active neurogenesis in human fetuses, and sox4 knockdown in Xenopus embryos diminishes brain and whole-body size. The SOX4 variants cluster in the highly conserved, SOX family-specific HMG domain, but each alters a different residue. In silico tools predict that each variant affects a distinct structural feature of this DNA-binding domain, and functional assays demonstrate that these SOX4 proteins carrying these variants are unable to bind DNA in vitro and transactivate SOX reporter genes in cultured cells. These variants are not found in the gnomAD database of individuals with presumably normal development, but 12 other SOX4 HMG-domain missense variants are recorded and all demonstrate partial to full activity in the reporter assay. Taken together, these findings point to specific SOX4 HMG-domain missense variants as the cause of a characteristic human neurodevelopmental disorder associated with mild facial and digital dysmorphism.


Subject(s)
Abnormalities, Multiple/genetics , Mutation, Missense/genetics , Neurodevelopmental Disorders/genetics , SOXC Transcription Factors/genetics , Amino Acid Sequence , Animals , Child , Child, Preschool , Coffin-Lowry Syndrome/genetics , Cohort Studies , Conserved Sequence , DNA/genetics , DNA/metabolism , Female , HMG-Box Domains/genetics , Heterozygote , Humans , Male , SOX Transcription Factors/chemistry , SOX Transcription Factors/genetics , SOXC Transcription Factors/chemistry , SOXC Transcription Factors/metabolism , Transcriptional Activation , Xenopus/anatomy & histology , Xenopus/embryology , Xenopus/genetics , Xenopus Proteins/chemistry , Xenopus Proteins/genetics
6.
FEMS Microbiol Lett ; 365(12)2018 06 01.
Article in English | MEDLINE | ID: mdl-29617942

ABSTRACT

Ustilago esculenta, an obligate parasite of Zizania latifolia, is a typical dimorphic fungus which induces host stem swelling and inhibits host inflorescence development, but is not found in host leaves. Previous studies have shown that dimorphic switching is essential for fungal pathogenicity and is regulated by protein kinase A and mitogen-activated protein kinase (MAPK) signaling pathways that are integrated by Prf1 in Ustilago maydis. In this study we identified a Prf1 homolog in U. esculenta, designated UePrf1, encoding 830 amino acids with a conserved high mobility group domain located between amino acids 124 and 195. UePrf1 was upregulated during the mating process, which induces dimorphism in U. esculenta. In vitro, UePrf1 mutants showed defects in the mating process, including cell fusion and hyphal growth. UePrf1 mutants also show reduced expression of a genes, even during the cell fusion process. Additionally, the defect in hyphal growth of the UeKpp2 and UeKpp6 mutants (MAPK signaling pathway mutants) was partially counteracted by UePrf1 overexpression, along with induced b gene expression. These results provide evidence that UePrf1 is a key factor coordinating dimorphism in U. esculenta and suggest a conserved role for UePrf1 in the regulation of the a and b genes.


Subject(s)
Fungal Proteins/genetics , Ustilago/genetics , Cloning, Molecular , Fungal Proteins/isolation & purification , Genes, Mating Type, Fungal/genetics , HMG-Box Domains/genetics , Host-Pathogen Interactions/genetics , Hyphae/genetics , Hyphae/growth & development , Mitogen-Activated Protein Kinases/genetics , Mutation , Plant Diseases/microbiology , Transcription Factors/genetics , Ustilago/growth & development , Ustilago/pathogenicity
7.
Sci Rep ; 8(1): 5156, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29581456

ABSTRACT

For decades, outbred guinea pigs (GP) have been used as research models. Various past research studies using guinea pigs used measures that, unknown at the time, may be sex-dependent, but from which today, archival tissues may be all that remain. We aimed to provide a protocol for sex-typing archival guinea pig tissue, whereby past experiments could be re-evaluated for sex effects. No PCR sex-genotyping protocols existed for GP. We found that published sequence of the GP Sry gene differed from that in two separate GP stocks. We used sequences from other species to deduce PCR primers for Sry. After developing a genomic DNA extraction for archival, fixed, decalcified, immunolabeled, guinea pig cochlear half-turns, we used a multiplex assay (Y-specific Sry; X-specific Dystrophin) to assign sex to tissue as old as 3 years. This procedure should allow reevaluation of prior guinea pig studies in various research areas for the effects of sex on experimental outcomes.


Subject(s)
Cochlea , Genes, sry/genetics , Genotype , Genotyping Techniques/methods , Guinea Pigs/genetics , Multiplex Polymerase Chain Reaction/methods , Tissue Banks , Amino Acid Sequence , Animals , Cloning, Molecular , DNA/isolation & purification , DNA Primers , Dystrophin/genetics , HMG-Box Domains/genetics , Immunohistochemistry , Sex Factors
8.
PLoS Genet ; 13(3): e1006622, 2017 03.
Article in English | MEDLINE | ID: mdl-28278156

ABSTRACT

HMG-box proteins, including Sox/SRY (Sox) and TCF/LEF1 (TCF) family members, bind DNA via their HMG-box. This binding, however, is relatively weak and both Sox and TCF factors employ distinct mechanisms for enhancing their affinity and specificity for DNA. Here we report that Capicua (CIC), an HMG-box transcriptional repressor involved in Ras/MAPK signaling and cancer progression, employs an additional distinct mode of DNA binding that enables selective recognition of its targets. We find that, contrary to previous assumptions, the HMG-box of CIC does not bind DNA alone but instead requires a distant motif (referred to as C1) present at the C-terminus of all CIC proteins. The HMG-box and C1 domains are both necessary for binding specific TGAATGAA-like sites, do not function via dimerization, and are active in the absence of cofactors, suggesting that they form a bipartite structure for sequence-specific binding to DNA. We demonstrate that this binding mechanism operates throughout Drosophila development and in human cells, ensuring specific regulation of multiple CIC targets. It thus appears that HMG-box proteins generally depend on auxiliary DNA binding mechanisms for regulating their appropriate genomic targets, but that each sub-family has evolved unique strategies for this purpose. Finally, the key role of C1 in DNA binding also explains the fact that this domain is a hotspot for inactivating mutations in oligodendroglioma and other tumors, while being preserved in oncogenic CIC-DUX4 fusion chimeras associated to Ewing-like sarcomas.


Subject(s)
DNA/genetics , Drosophila Proteins/genetics , HMGB Proteins/genetics , High Mobility Group Proteins/genetics , Mutation , Neoplasms/genetics , Repressor Proteins/genetics , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Base Sequence , Binding Sites/genetics , DNA/metabolism , Drosophila/embryology , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , HEK293 Cells , HMG-Box Domains/genetics , HMGB Proteins/metabolism , High Mobility Group Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Microscopy, Confocal , Models, Genetic , Neoplasms/metabolism , Protein Binding , Repressor Proteins/metabolism , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
9.
Oncotarget ; 8(29): 46955-46968, 2017 Jul 18.
Article in English | MEDLINE | ID: mdl-28187450

ABSTRACT

The nucleocytoplasmic shuttling of SOX transcription factors play a crucial role in the regulation of SOX protein functions during development. In this study, we have demonstrated two nuclear localization signals in the HMG box of Eriocheir sinensis SOX14A and SOX14B. These two conserved nuclear localization signals mediate nuclear transport. The N-termini nuclear localization signal mediates the calmodulin-dependent pathway and the C-termini nuclear localization signal interacts with the importin-ß pathway. The targeted deletion of nuclear localization signals of SOX14A/B dramatically inhibits the nuclear accumulation. We have first time revealed a non-classic nuclear export signal in the HMG box of E. sinensis SOX14A/B proteins is responds to leptomycin B. E. sinensis SOX14A/B is transported from the nucleus to the cytoplasm via a CRM1-dependent nuclear export pathway. And E. sinensis SOX14A/B are not belong to the subgroup E SOX proteins. Furthermore, these findings could shed a light on the mechanisms involved in the nuclear export of SOX proteins. The imperfect nuclear export signal on other SOX proteins, rather than just those of the SOXE group, may also be functional for nuclear export.


Subject(s)
SOXB2 Transcription Factors/metabolism , Amino Acid Sequence , Animals , Brachyura , Cell Nucleus/metabolism , Cytoplasm/metabolism , HEK293 Cells , HMG-Box Domains/genetics , Humans , Karyopherins/metabolism , Models, Biological , Nuclear Localization Signals , Receptors, Cytoplasmic and Nuclear/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , SOXB2 Transcription Factors/chemistry , SOXB2 Transcription Factors/genetics , Exportin 1 Protein
10.
Biochim Biophys Acta Gene Regul Mech ; 1860(2): 256-269, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27871851

ABSTRACT

Ixr1 is a transcriptional factor involved in the response to hypoxia, which is also related to DNA repair. It binds to DNA through its two in-tandem high mobility group box (HMG-box) domains. Each function depends on recognition of different DNA structures, B-form DNA at specific consensus sequences for transcriptional regulation, or distorted DNA, like cisplatin-DNA adducts, for DNA repair. However, the contribution of the HMG-box domains in the Ixr1 protein to the formation of different protein-DNA complexes is poorly understood. We have biophysically and biochemically characterized these interactions with specific DNA sequences from the promoters regulated by Ixr1, or with cisplatin-DNA adducts. Both HMG-boxes are necessary for transcriptional regulation, and they are not functionally interchangeable. The in-tandem arrangement of their HMG-boxes is necessary for functional folding and causes sequential cooperative binding to specific DNA sequences, with HMG-box A showing a higher contribution to DNA binding and bending than the HMG-box B. Binding of Ixr1 HMG boxes to specific DNA sequences is entropy driven, whereas binding to platinated DNA is enthalpy driven for HMG-box A and entropy driven for HMG-box B. This is the first proof that HMG-box binding to different DNA structures is associated with predictable thermodynamic differences. Based on our study, we present a model to explain the dual function of Ixr1 in the regulation of gene expression and recognition of distorted DNA structures caused by cisplatin treatment.


Subject(s)
Cisplatin/metabolism , DNA Adducts/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Fungal/genetics , HMG-Box Domains/genetics , High Mobility Group Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription, Genetic/genetics , Amino Acid Sequence , DNA/metabolism , DNA Repair/genetics , Nucleic Acid Conformation , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Protein Folding , Saccharomyces cerevisiae/metabolism , Sequence Alignment , Thermodynamics
11.
Genes Cells ; 21(12): 1333-1352, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27860073

ABSTRACT

Hmo1, a member of HMGB family proteins in Saccharomyces cerevisiae, binds to and regulates the transcription of genes encoding ribosomal RNA and ribosomal proteins. The functional motifs of Hmo1 include two HMG-like motifs, box A and box B, and a C-terminal tail. To elucidate the molecular roles of the HMG-like boxes in DNA binding in vivo, we analyzed the DNA-binding activity of various Hmo1 mutants using ChIP or reporter assays that enabled us to conveniently detect Hmo1 binding to the promoter of RPS5, a major target gene of Hmo1. Our mutational analyses showed that box B is a bona fide DNA-binding motif and that it also plays other important roles in cell growth. However, box A, especially its first α-helix, contributes to DNA binding of Hmo1 by inducing self-assembly of Hmo1. Intriguingly, box A mediated formation of oligomers of more than two proteins on DNA in vivo. Furthermore, duplication of the box B partially alleviates the requirement for box A. These findings suggest that the principal role of box A is to assemble multiple box B in the appropriate orientation, thereby stabilizing the binding of Hmo1 to DNA and nucleating specific chromosomal architecture on its target genes.


Subject(s)
DNA, Fungal/metabolism , HMG-Box Domains , High Mobility Group Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HMG-Box Domains/genetics , High Mobility Group Proteins/chemistry , High Mobility Group Proteins/genetics , Mutation , Protein Binding , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
12.
PLoS One ; 11(8): e0161432, 2016.
Article in English | MEDLINE | ID: mdl-27532129

ABSTRACT

Group E members of the SOX transcription factor family include SOX8, SOX9, and SOX10. Preceding the high mobility group (HMG) domain in each of these proteins is a thirty-eight amino acid region that supports the formation of dimers on promoters containing tandemly inverted sites. The purpose of this study was to obtain new structural insights into how the dimerization region functions with the HMG domain. From a mutagenic scan of the dimerization region, the most essential amino acids of the dimerization region were clustered on the hydrophobic face of a single, predicted amphipathic helix. Consistent with our hypothesis that the dimerization region directly contacts the HMG domain, a peptide corresponding to the dimerization region bound a preassembled HMG-DNA complex. Sequence conservation among Group E members served as a basis to identify two surface exposed amino acids in the HMG domain of SOX9 that were necessary for dimerization. These data were combined to make a molecular model that places the dimerization region of one SOX9 protein onto the HMG domain of another SOX9 protein situated at the opposing site of a tandem promoter. The model provides a detailed foundation for assessing the impact of mutations on SOX Group E transcription factors.


Subject(s)
DNA-Binding Proteins/metabolism , HMG-Box Domains/genetics , Models, Molecular , Protein Multimerization/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Binding Sites/genetics , Dimerization , Electrophoretic Mobility Shift Assay , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Docking Simulation , Promoter Regions, Genetic
13.
J. physiol. biochem ; 71(3): 359-372, sept. 2015.
Article in English | IBECS | ID: ibc-142435

ABSTRACT

The expression of the proinflammatory cytokine high-mobility group box-1 (HMGB1) is upregulated in epiretinal membranes and vitreous fluid from patients with proliferative diabetic retinopathy (PDR) and in the diabetic retina. We hypothesized that a novel mechanism exists where HMGB1 and NADPH oxidase (Nox)-derived reactive oxygen species (ROS) are mutually enhanced in the diabetic retina, which may be a novel mechanism for promoting upregulation of retinal apoptotic markers induced by diabetes. Vitreous samples from 48 PDR and 34 nondiabetic patients, retinas from 1-month diabetic rats and from normal rats intravitreally injected with HMGB1 and human retinal microvascular endothelial cells (HRMEC) stimulated with HMGB1 were studied by enzyme-linked immunosorbent and spectrophotometric assays, Western blot analysis, RT-PCR, and immunofluorescence. We also studied the effect of the HMGB1 inhibitor glycyrrhizin and apocynin on diabetes-induced biochemical changes in the retinas of rats (n = 5-7 in each groups). HMGB1 and the oxidative stress marker protein carbonyl content levels in the vitreous fluid from PDR patients were significantly higher than in controls (p = 0.021; p = 0.005, respectively). There was a significant positive correlation between vitreous fluid levels of HMGB1 and the levels of protein carbonyl content (r = 0.62, p = 0.001). HMGB1 enhanced interleukin-1β, ROS, Nox2, poly (ADP-ribose) polymerase (PARP)-1, and cleaved caspase-3 production by HRMEC. Diabetes and intravitreal injection of HMGB1 in normal rats induced significant upregulation of ROS, Nox2, PARP-1, and cleaved caspase-3 in the retina. Constant glycyrrhizin and apocynin intake from onset of diabetes did not affect the metabolic status of the diabetic rats, but restored these increased mediators to control values. The results of this study suggest that there is a mutual enhancement between HMGB1 and Nox-derived ROS in the diabetic retina, which may promote diabetes-induced upregulation of retinal apoptotic markers


Subject(s)
Animals , Rats , Diabetes Mellitus/physiopathology , Apoptosis/physiology , HMG-Box Domains/genetics , NADH, NADPH Oxidoreductases/genetics , Biomarkers/analysis , Retina/ultrastructure , Disease Models, Animal
14.
Biochim Biophys Acta ; 1849(6): 677-87, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25748361

ABSTRACT

The high-mobility group (HMG) domain containing proteins regulate transcription, DNA replication and recombination. They adopt L-shaped folds and are structure-specific DNA binding motifs. Here, I define the L-motif super-family that consists of DNA-binding HMG-box proteins and the L-motif of the histone mRNA binding domain of stem-loop binding protein (SLBP). The SLBP L-motif and HMG-box domains adopt similar L-shaped folds with three α-helices and two or three small hydrophobic cores that stabilize the overall fold, but have very different and distinct modes of nucleic acid recognition. A comparison of the structure, dynamics, protein-protein and nucleic acid interactions, and regulation by PTMs of the SLBP and the HMG-box L-motifs reveals the versatile and diverse modes by which L-motifs utilize their surfaces for structure-specific recognition of nucleic acids to regulate gene expression.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation , HMG-Box Domains/genetics , Nuclear Proteins/genetics , RNA-Binding Proteins/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Amino Acid Motifs/genetics , DNA-Binding Proteins/chemistry , Humans , Inverted Repeat Sequences/genetics , Nuclear Proteins/chemistry , Nucleic Acid Conformation , Phosphorylation , Protein Conformation , Protein Folding , RNA-Binding Proteins/chemistry , mRNA Cleavage and Polyadenylation Factors/chemistry
15.
PLoS Genet ; 10(8): e1004509, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25144371

ABSTRACT

The Wnt/ß-catenin signaling pathway plays many important roles in animal development, tissue homeostasis and human disease. Transcription factors of the TCF family mediate many Wnt transcriptional responses, promoting signal-dependent activation or repression of target gene expression. The mechanism of this specificity is poorly understood. Previously, we demonstrated that for activated targets in Drosophila, TCF/Pangolin (the fly TCF) recognizes regulatory DNA through two DNA binding domains, with the High Mobility Group (HMG) domain binding HMG sites and the adjacent C-clamp domain binding Helper sites. Here, we report that TCF/Pangolin utilizes a similar bipartite mechanism to recognize and regulate several Wnt-repressed targets, but through HMG and Helper sites whose sequences are distinct from those found in activated targets. The type of HMG and Helper sites is sufficient to direct activation or repression of Wnt regulated cis-regulatory modules, and protease digestion studies suggest that TCF/Pangolin adopts distinct conformations when bound to either HMG-Helper site pair. This repressive mechanism occurs in the fly lymph gland, the larval hematopoietic organ, where Wnt/ß-catenin signaling controls prohemocytic differentiation. Our study provides a paradigm for direct repression of target gene expression by Wnt/ß-catenin signaling and allosteric regulation of a transcription factor by DNA.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , HMG-Box Domains/genetics , Hematopoietic System/metabolism , Repressor Proteins/genetics , Animals , Binding Sites , Drosophila Proteins/metabolism , Drosophila melanogaster , Humans , Lymph/metabolism , Repressor Proteins/metabolism , Transcriptional Activation/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics
16.
PLoS Genet ; 10(2): e1004133, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24516405

ABSTRACT

Regulation of gene expression by signaling pathways often occurs through a transcriptional switch, where the transcription factor responsible for signal-dependent gene activation represses the same targets in the absence of signaling. T-cell factors (TCFs) are transcription factors in the Wnt/ß-catenin pathway, which control numerous cell fate specification events in metazoans. The TCF transcriptional switch is mediated by many co-regulators that contribute to repression or activation of Wnt target genes. It is typically assumed that DNA recognition by TCFs is important for target gene location, but plays no role in the actual switch. TCF/Pangolin (the fly TCF) and some vertebrate TCF isoforms bind DNA through two distinct domains, a High Mobility Group (HMG) domain and a C-clamp, which recognize DNA motifs known as HMG and Helper sites, respectively. Here, we demonstrate that POP-1 (the C. elegans TCF) also activates target genes through HMG and Helper site interactions. Helper sites enhanced the ability of a synthetic enhancer to detect Wnt/ß-catenin signaling in several tissues and revealed an unsuspected role for POP-1 in regulating the C. elegans defecation cycle. Searching for HMG-Helper site clusters allowed the identification of a new POP-1 target gene active in the head muscles and gut. While Helper sites and the C-clamp are essential for activation of worm and fly Wnt targets, they are dispensable for TCF-dependent repression of targets in the absence of Wnt signaling. These data suggest that a fundamental change in TCF-DNA binding contributes to the transcriptional switch that occurs upon Wnt stimulation.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/genetics , Gene Expression Regulation , High Mobility Group Proteins/metabolism , Repressor Proteins/metabolism , Animals , Binding Sites , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , DNA-Binding Proteins/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , HMG-Box Domains/genetics , High Mobility Group Proteins/genetics , Nucleotide Motifs/genetics , Protein Binding , Repressor Proteins/genetics , Signal Transduction/genetics , Wnt Signaling Pathway/genetics
17.
PLoS Genet ; 9(7): e1003642, 2013.
Article in English | MEDLINE | ID: mdl-23935511

ABSTRACT

High-mobility group (HMG) B proteins are eukaryotic DNA-binding proteins characterized by the HMG-box functional motif. These transcription factors play a pivotal role in global genomic functions and in the control of genes involved in specific developmental or metabolic pathways. The filamentous ascomycete Podospora anserina contains 12 HMG-box genes. Of these, four have been previously characterized; three are mating-type genes that control fertilization and development of the fruit-body, whereas the last one encodes a factor involved in mitochondrial DNA stability. Systematic deletion analysis of the eight remaining uncharacterized HMG-box genes indicated that none were essential for viability, but that seven were involved in the sexual cycle. Two HMG-box genes display striking features. PaHMG5, an ortholog of SpSte11 from Schizosaccharomyces pombe, is a pivotal activator of mating-type genes in P. anserina, whereas PaHMG9 is a repressor of several phenomena specific to the stationary phase, most notably hyphal anastomoses. Transcriptional analyses of HMG-box genes in HMG-box deletion strains indicated that PaHMG5 is at the hub of a network of several HMG-box factors that regulate mating-type genes and mating-type target genes. Genetic analyses revealed that this network also controls fertility genes that are not regulated by mating-type transcription factors. This study points to the critical role of HMG-box members in sexual reproduction in fungi, as 11 out of 12 members were involved in the sexual cycle in P. anserina. PaHMG5 and SpSte11 are conserved transcriptional regulators of mating-type genes, although P. anserina and S. pombe diverged 550 million years ago. Two HMG-box genes, SOX9 and its upstream regulator SRY, also play an important role in sex determination in mammals. The P. anserina and S. pombe mating-type genes and their upstream regulatory factor form a module of HMG-box genes analogous to the SRY/SOX9 module, revealing a commonality of sex regulation in animals and fungi.


Subject(s)
DNA-Binding Proteins/genetics , Genes, Mating Type, Fungal , High Mobility Group Proteins/genetics , Podospora/genetics , DNA-Binding Proteins/metabolism , Fertilization/genetics , Gene Expression Regulation, Fungal , HMG-Box Domains/genetics , High Mobility Group Proteins/metabolism , Multigene Family , Podospora/physiology , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Schizosaccharomyces/genetics , Sequence Deletion , Transcription Factors/genetics , Transcription Factors/metabolism
18.
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
19.
Dig Dis Sci ; 58(11): 3198-206, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23861108

ABSTRACT

BACKGROUND AND OBJECTIVES: Previous studies have shown that betaine prevents alcohol-induced liver injury and improves liver function. The purpose of this study was to investigate the hepatoprotective effects of betaine on nonalcoholic fatty liver disease (NAFLD) and to observe changes of HMGB1/TLR4 signaling. METHODS: Thirty rats were randomly divided into control, model, and betaine groups. The rats in the model and betaine groups were fed a high-fat diet for 12 weeks to induce an animal model of NAFLD. The rats in the betaine group were then intragastrically administered betaine solution at a dose of 400 mg/kg per day for four weeks. Liver histology was examined. Serum levels of ALT, AST, TC, TG, HDL-C, LDL-C, FFA, HMGB1, NF-κB, TLR4, and tHcy were determined and intrahepatic TC, TG, and Hcy levels were assayed. mRNA expression and protein levels of HMGB1, NF-κB, and TLR4 in liver tissue were also determined. RESULTS: Compared with the control group, rats in the model group developed severe liver injury, accompanied by significant increases in serum levels of ALT, AST, TC, TG, LDL-C, FFA, HMGB1, NF-κB, and TLR4, intrahepatic TC, TG, and Hcy content, histological scores for steatosis, inflammation, and necrosis, and mRNA expression and protein levels of HMGB1, NF-κB, and TLR4, and a significant decrease in serum HDL-C (P < 0.05). Compared with the model group, all these indicators were significantly improved by administration of betaine (P < 0.05). CONCLUSIONS: Betaine effectively protects against high-fat-diet-induced NAFLD and improves liver function; the mechanism is probably related to inhibition of HMGB1/TLR4 signaling pathways.


Subject(s)
Betaine/pharmacology , Chemical and Drug Induced Liver Injury/prevention & control , Dietary Fats/adverse effects , Gene Expression Regulation/physiology , HMG-Box Domains/physiology , Toll-Like Receptor 4/metabolism , Animals , Cytokines/genetics , Cytokines/metabolism , Fatty Liver/prevention & control , Female , HMG-Box Domains/genetics , Lipid Metabolism , Liver/drug effects , Liver/enzymology , Liver/metabolism , Non-alcoholic Fatty Liver Disease , Rats , Rats, Sprague-Dawley , Specific Pathogen-Free Organisms , Toll-Like Receptor 4/genetics , Weight Gain
20.
Genetika ; 48(1): 89-96, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22567858

ABSTRACT

The Sox (SRY-related high-mobility-group box) family of genes shares a conserved HMG box and is involved in a diverse range of developmental processes and sex determination in vertebrates. Twenty Sox genes are present in the genomes of humans and mice, but far less is known about the Sox gene family in reptiles. Using two pairs of highly degenerate primers designed from a multiple alignment of Sox amino acid sequences in several species, different positive clones were obtained from male and female Eremias multiocellata, a viviparous lizard which is subject to TSD (temperature-dependent sex determination). These clones were sequenced and identified. They are members of the SoxB (Sox2, Sox14), SoxC (Sox11, Sox12) and SoxE (Sox9a, Sox9b, Sox10) groups. No sex-specific differences were observed. Based on the amino acid sequence similarities, the phylogenetic analysis was carried out and these genes clustered with their orthologues. In addition, we found the gene duplication in E. multiocellata, it may be a mechanism to produce new functional genes.


Subject(s)
Lizards/genetics , SOXB1 Transcription Factors/genetics , SOXC Transcription Factors/genetics , SOXE Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Female , HMG-Box Domains/genetics , Humans , Male , Mice , Molecular Sequence Data , Phylogeny , SOXB1 Transcription Factors/isolation & purification , SOXC Transcription Factors/isolation & purification , SOXE Transcription Factors/isolation & purification , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sex Determination Processes
SELECTION OF CITATIONS
SEARCH DETAIL
...