Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 35
1.
Genetics ; 224(4)2023 08 09.
Article En | MEDLINE | ID: mdl-37067863

TWIST1 is a basic helix-loop-helix (bHLH) transcription factor in humans that functions in mesoderm differentiation. TWIST1 primarily regulates genes as a transcriptional repressor often through TWIST-Box domain-mediated protein-protein interactions. The TWIST-Box also can function as an activation domain requiring 3 conserved, equidistant amino acids (LXXXFXXXR). Autosomal dominant mutations in TWIST1, including 2 reported in these conserved amino acids (F187L and R191M), lead to craniofacial defects in Saethre-Chotzen syndrome (SCS). Caenorhabditis elegans has a single TWIST1 homolog, HLH-8, that functions in the differentiation of the muscles responsible for egg laying and defecation. Null alleles in hlh-8 lead to severely egg-laying defective and constipated animals due to defects in the corresponding muscles. TWIST1 and HLH-8 share sequence identity in their bHLH regions; however, the domain responsible for the transcriptional activity of HLH-8 is unknown. Sequence alignment suggests that HLH-8 has a TWIST-Box LXXXFXXXR motif; however, its function also is unknown. CRISPR/Cas9 genome editing was utilized to generate a domain deletion and several missense mutations, including those analogous to SCS patients, in the 3 conserved HLH-8 amino acids to investigate their functional role. The TWIST-Box alleles did not phenocopy hlh-8 null mutants. The strongest phenotype detected was a retentive (Ret) phenotype with late-stage embryos in the hermaphrodite uterus. Further, GFP reporters of HLH-8 downstream target genes (arg-1::gfp and egl-15::gfp) revealed tissue-specific, target-specific, and allele-specific defects. Overall, the TWIST-Box in HLH-8 is partially required for the protein's transcriptional activity, and the conserved amino acids contribute unequally to the domain's function.


Acrocephalosyndactylia , Caenorhabditis elegans , Animals , Female , Humans , Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Mutation , Transcription Factors/genetics , Twist-Related Protein 1/genetics , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/metabolism
2.
Int J Biol Macromol ; 194: 24-31, 2022 Jan 01.
Article En | MEDLINE | ID: mdl-34863830

Twist1 promote the bypass of p53 response by interacting with p53 and facilitating its MDM2-mediated degradation. We reasoned that reagents able to interfere with the p53:Twist1 complex might alleviate Twist1 inhibitory effect over p53, thus representing potential therapeutic tools in p53 wild type tumors. From a pre-immune library of llama nanobodies (VHH), we isolated binders targeting the p53 C-terminal region (p53-CTD) involved in the interaction with Twist1 by using recombinant Twist1 as an epitope-specific competitor during elution. Positive hits were validated by proving their capacity to immunoprecipitate p53 and to inhibit Twist1:p53 binding in vitro. Molecular modeling confirmed a preferential docking of positive hits with p53-CTD. D11 VHH activity was validated in human cell models, succeeded in immunoprecipitating endogenous p53 and, similarly to Twist1 knock-down, interfered with p53 turnover, p53 phosphorylation at Serine 392 and affected cell viability. Despite the limited functional effect determined by D11 expression in target cells, our results provide the proof of principle that nanobodies ectopically expressed within a cell, have the capacity to target the assembly of the pro-tumorigenic Twist1:p53 complex. These results disclose novel tools for dissecting p53 biology and lay down the grounds for the development of innovative targeted therapeutic approaches.


Single-Domain Antibodies/chemistry , Tumor Suppressor Protein p53/chemistry , Twist-Related Protein 1/chemistry , Binding, Competitive , Cell Line , Epitopes/chemistry , Epitopes/immunology , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Recombinant Proteins , Structure-Activity Relationship , Tumor Suppressor Protein p53/metabolism , Twist-Related Protein 1/metabolism
3.
Mol Cell Biol ; 40(11)2020 05 14.
Article En | MEDLINE | ID: mdl-32179550

The extensive array of basic helix-loop-helix (bHLH) transcription factors and their combinations as dimers underpin the diversity of molecular function required for cell type specification during embryogenesis. The bHLH factor TWIST1 plays pleiotropic roles during development. However, which combinations of TWIST1 dimers are involved and what impact each dimer imposes on the gene regulation network controlled by TWIST1 remain elusive. In this work, proteomic profiling of human TWIST1-expressing cell lines and transcriptome analysis of mouse cranial mesenchyme have revealed that TWIST1 homodimers and heterodimers with TCF3, TCF4, and TCF12 E-proteins are the predominant dimer combinations. Disease-causing mutations in TWIST1 can impact dimer formation or shift the balance of different types of TWIST1 dimers in the cell, which may underpin the defective differentiation of the craniofacial mesenchyme. Functional analyses of the loss and gain of TWIST1-E-protein dimer activity have revealed previously unappreciated roles in guiding lineage differentiation of embryonic stem cells: TWIST1-E-protein heterodimers activate the differentiation of mesoderm and neural crest cells, which is accompanied by the epithelial-to-mesenchymal transition. At the same time, TWIST1 homodimers maintain the stem cells in a progenitor state and block entry to the endoderm lineage.


Cell Differentiation , Nuclear Proteins/metabolism , Protein Multimerization , Twist-Related Protein 1/metabolism , Animals , Cell Line , Dogs , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Developmental , Humans , Madin Darby Canine Kidney Cells , Mesoderm/cytology , Mesoderm/metabolism , Mice, Inbred C57BL , Mutation , Neural Crest/cytology , Neural Crest/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Transcriptome , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/genetics
4.
Sci Rep ; 10(1): 2501, 2020 02 12.
Article En | MEDLINE | ID: mdl-32051525

Twist1 encodes a basic helix-loop-helix transcription factor (TF), which forms homodimer or heterodimer with other TFs, like E2A, to regulate target genes' expression. Mutations in TWIST1 are associated with Saethre-Chotzen syndrome (SCS), a rare congenital disorder characterized with osteogenesis abnormalities. However, how dysfunction of TWIST1 leads to SCS is still largely unknown. Here, using an unbiased ENU-induced mutagenesis screening, we identified a novel Twist1 mutation and the mutant mouse phenocopies some features of SCS in a dominant manner. Physically, our mutation p.F191S lies at the edge of a predicted α-helix in Twist1 transactivation (TA) domain. Adjacent to F191, a consecutive three-residue (AFS) has been hit by 3 human and 2 mouse disease-associated mutations, including ours. Unlike previously reported mouse null and p.S192P alleles that lead to hindlimb polydactyly with incomplete penetrance but a severe craniofacial malformation, our p.F191S causes the polydactyly (84.2% bilateral and 15.8% unilateral) with complete penetrance but a mild craniofacial malformation. Consistent with the higher penetrance, p.F191S has stronger impairment on E2A-dependent transcription than p.S192P. Although human p.A186T and mouse p.S192P disease mutations are adjacent to ours, these three mutations function differently to impair the E2A-dependent transcription. Unlike p.A186T and p.S192S that disturb local protein conformation and unstabilize the mutant proteins, p.F191S keeps the mutant protein stable and its interaction with E2A entire. Therefore, we argue that p.F191S we identified acts in a dominant-negative manner to impair E2A-dependent transcription and to cause the biological consequences. In addition, the mutant mouse we provided here could be an additional and valuable model for better understanding the disease mechanisms underlying SCS caused by TWIST1 dysfunction.


Mutation , Penetrance , Polydactyly/genetics , Twist-Related Protein 1/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Ethylnitrosourea/toxicity , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mutagens/toxicity , Protein Domains , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/metabolism
5.
Medicine (Baltimore) ; 98(52): e18449, 2019 Dec.
Article En | MEDLINE | ID: mdl-31876727

Twist and E-cadherin are crucial for the development of different types of cancer; however, their clinical significance in adenocarcinoma of the gastroesophageal junction (AGE) remains unknown. Here, we investigated the correlation between the expression of Twist and E-cadherin and their impact on the clinical outcomes and prognosis of patients with AGE and proximal gastric carcinoma (PGC).Using immunohistochemistry, we determined the expression of Twist and E-cadherin in the tissue samples of patients with AGE and PGC. The correlation of the expression of Twist and E-cadherin with the clinicopathological factors was assessed by using the chi-square test, Fisher exact test, and non-parametric Mann-Whitney U test. The Kaplan-Meier method along with the log-rank test and Cox proportional-hazards model were used to evaluate the correlation of Twist and E-cadherin expression with the overall survival (OS) of patients.Overall, 94 patients with AGE (n = 45, 47.87%) or PGC (n = 49, 52.13%) who underwent primary tumor resection were included in this study. The median follow-up period was 40.5 months. We observed a significant difference in the smoking status (P < .001) and differentiation grade (P = .004) between patients with AGE and PGC. There was a significant association of a high Twist expression with T stage (only in PGC, P = .008), lymph node metastasis (AGE, P = .075; PGC, P = .051), and advanced pathological stages (AGE, P = .019; PGC, P = .006). A low E-cadherin expression showed similar results; however, it was not significantly associated with the advanced pathological stages of AGE (P = .372). A low E-cadherin expression was significantly associated with a low differentiation grade of AGE (P = .002). In addition, a significant inverse relationship was observed between Twist and E-cadherin expression. The Kaplan-Meier survival analysis and Cox regression analysis revealed that a high Twist expression and low E-cadherin expression were independent prognostic factors for short OS of patients with AGE or PGC.A high Twist expression or low E-cadherin expression was associated with unfavorable clinicopathological factors and independently predicted short OS of patients with AGE or PGC.


Adenocarcinoma/metabolism , Cadherins/metabolism , Carcinoma/metabolism , Esophagogastric Junction/metabolism , Nuclear Proteins/metabolism , Stomach Neoplasms/metabolism , Twist-Related Protein 1/metabolism , Adenocarcinoma/chemistry , Adenocarcinoma/diagnosis , Adult , Aged , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/metabolism , Cadherins/analysis , Carcinoma/chemistry , Carcinoma/diagnosis , Esophagogastric Junction/chemistry , Female , Humans , Male , Middle Aged , Nuclear Proteins/chemistry , Prognosis , Stomach Neoplasms/chemistry , Stomach Neoplasms/diagnosis , Twist-Related Protein 1/chemistry
6.
Genes Dev ; 33(11-12): 626-640, 2019 06 01.
Article En | MEDLINE | ID: mdl-30975722

Rhabdomyosarcoma (RMS) is an aggressive pediatric cancer composed of myoblast-like cells. Recently, we discovered a unique muscle progenitor marked by the expression of the Twist2 transcription factor. Genomic analyses of 258 RMS patient tumors uncovered prevalent copy number amplification events and increased expression of TWIST2 in fusion-negative RMS. Knockdown of TWIST2 in RMS cells results in up-regulation of MYOGENIN and a decrease in proliferation, implicating TWIST2 as an oncogene in RMS. Through an inducible Twist2 expression system, we identified Twist2 as a reversible inhibitor of myogenic differentiation with the remarkable ability to promote myotube dedifferentiation in vitro. Integrated analysis of genome-wide ChIP-seq and RNA-seq data revealed the first dynamic chromatin and transcriptional landscape of Twist2 binding during myogenic differentiation. During differentiation, Twist2 competes with MyoD at shared DNA motifs to direct global gene transcription and repression of the myogenic program. Additionally, Twist2 shapes the epigenetic landscape to drive chromatin opening at oncogenic loci and chromatin closing at myogenic loci. These epigenetic changes redirect MyoD binding from myogenic genes toward oncogenic, metabolic, and growth genes. Our study reveals the dynamic interplay between two opposing transcriptional regulators that control the fate of RMS and provides insight into the molecular etiology of this aggressive form of cancer.


Carcinogenesis , Muscle Development , MyoD Protein/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Cells, Cultured , Chromatin Assembly and Disassembly , DNA/metabolism , Epithelial-Mesenchymal Transition , Gene Amplification , Gene Expression Regulation, Neoplastic , HEK293 Cells , Helix-Loop-Helix Motifs , Humans , MyoD Protein/chemistry , Myoblasts/metabolism , Nuclear Proteins/genetics , Repressor Proteins/chemistry , Twist-Related Protein 1/chemistry
7.
J Biol Chem ; 294(13): 5082-5093, 2019 03 29.
Article En | MEDLINE | ID: mdl-30733340

Twist1 is a basic helix-loop-helix transcription factor that plays a key role in embryonic development, and its expression is down-regulated in adult cells. However, Twist1 is highly expressed during cancer development, conferring a proliferative, migratory, and invasive phenotype to malignant cells. Twist1 expression can be regulated post-translationally by phosphorylation or ubiquitination events. We report in this study a previously unknown and relevant Twist1 phosphorylation site that controls its stability. To identify candidate phosphorylation sites in Twist1, we first conducted an in silico analysis of the Twist1 protein, which yielded several potential sites. Because most of these sites were predicted to be phosphorylated by protein kinase C (PKC), we overexpressed PKCα in several cell lines and found that it phosphorylates Twist1 on Ser-144. Using a combination of immunoblotting, immunoprecipitation, protein overexpression, and CRISPR/Cas9-mediated PKCα knockout experiments, we observed that PKCα-mediated Twist1 phosphorylation at Ser-144 inhibits Twist1 ubiquitination and consequently stabilizes it. These results provide evidence for a direct association between PKCα and Twist1 and yield critical insights into the PKCα/Twist1 signaling axis that governs cancer aggressiveness.


Nuclear Proteins/metabolism , Protein Kinase C-alpha/metabolism , Twist-Related Protein 1/metabolism , Ubiquitination , Epithelial-Mesenchymal Transition , HEK293 Cells , Humans , Models, Molecular , Nuclear Proteins/chemistry , Phosphorylation , Protein Interaction Domains and Motifs , Protein Stability , Twist-Related Protein 1/chemistry
8.
Am J Med Genet A ; 176(12): 2777-2780, 2018 12.
Article En | MEDLINE | ID: mdl-30450715

The TWIST family is a group of highly conserved basic helix-loop-helix transcription factors. In humans, TWIST1 haploinsufficiency causes Saethre-Chotzen syndrome, which is characterized by craniosynostosis. Heterozygous localized TWIST1 and TWIST2 basic domain substitutions exert antimorphic effects to cause Sweeney-Cox syndrome, Barber-Say syndrome, and ablepharon-macrostomia syndrome, respectively. Sweeney-Cox syndrome, Barber-Say syndrome, and ablepharon-macrostomia syndrome share the facial features of ablepharon, hypertelorism, underdevelopment of the eyelids, and cheek pads adjacent to the corners of the mouth. Existence of phenotypic overlap between Saethre-Chotzen syndrome and Sweeney-Cox syndrome remains unknown. Herein, we document a male infant with the distinctive facial features of ablepharon, hypertelorism, cheek pads adjacent to the corners of the mouth, and bilateral coronal suture craniosynostosis who had a de novo heterozygous mutation in the basic domain of TWIST1, that is, c.351C>G p.Glu117Asp. The pathogenicity of this variant was supported by in silico and in vivo evidence. Our review showed that Sweeney-Cox syndrome appears to share many characteristics with Barber-Say syndrome and ablepharon-macrostomia syndrome except for craniosynostosis, which is a cardinal feature of Saethre-Chotzen syndrome. An amino acid substitution from Glu117 to Asp, both of which are the sole members of negatively charged amino acids, resulted in a prototypic Sweeney-Cox syndrome phenotype. This suggests that any amino acid substitutions at Glu117 would likely lead to the Sweeney-Cox syndrome phenotype or lethality. The present observation suggests that a localized TWIST1 basic domain substitution, that is, p.Glu117Asp, in TWIST1 may exert a mild antimorphic effect similar to that of haploinsufficiency, leading to craniosynostosis and ablepharon.


Craniosynostoses/diagnosis , Craniosynostoses/genetics , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Nuclear Proteins/genetics , Protein Domains/genetics , Twist-Related Protein 1/genetics , Alleles , Amino Acid Substitution , Facies , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Infant, Newborn , Male , Nuclear Proteins/chemistry , Syndrome , Tomography, Spiral Computed , Twist-Related Protein 1/chemistry
9.
BMC Struct Biol ; 17(1): 6, 2017 05 18.
Article En | MEDLINE | ID: mdl-28521820

BACKGROUND: The bHLH transcription factor TWIST1 plays a key role in the embryonic development and in tumorigenesis. Some loss-of-function mutations of the TWIST1 gene have been shown to cause an autosomal dominant craniosynostosis, known as the Saethre-Chotzen syndrome (SCS). Although the functional impacts of many TWIST1 mutations have been experimentally reported, little is known on the molecular mechanisms underlying their loss-of-function. In a previous study, we highlighted the predictive value of in silico molecular dynamics (MD) simulations in deciphering the molecular function of TWIST1 residues. RESULTS: Here, since the substitution of the arginine 154 amino acid by a glycine residue (R154G) is responsible for the SCS phenotype and the substitution of arginine 154 by a proline experimentally decreases the dimerizing ability of TWIST1, we investigated the molecular impact of this point mutation using MD approaches. Consistently, MD simulations highlighted a clear decrease in the stability of the α-helix during the dimerization of the mutated R154P TWIST1/E12 dimer compared to the wild-type TE complex, which was further confirmed in vitro using immunoassays. CONCLUSIONS: Our study demonstrates that MD simulations provide a structural explanation for the loss-of-function associated with the SCS TWIST1 mutation and provides a proof of concept of the predictive value of these MD simulations. This in silico methodology could be used to determine reliable pharmacophore sites, leading to the application of docking approaches in order to identify specific inhibitors of TWIST1 complexes.


Molecular Dynamics Simulation , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Point Mutation , Transcription Factor 3/chemistry , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/genetics , Amino Acid Sequence , Animals , Arginine/chemistry , Arginine/genetics , Computer Simulation , Crystallography, X-Ray , Glycine/chemistry , Glycine/genetics , Humans , Mice , Phosphorylation , Protein Conformation , Protein Multimerization , Sequence Homology , Transcription Factor 3/genetics
10.
BMC Cancer ; 17(1): 184, 2017 03 10.
Article En | MEDLINE | ID: mdl-28283022

BACKGROUND: Most cancer deaths result from tumor cells that have metastasized beyond their tissue of origin, or have developed drug resistance. Across many cancer types, patients with advanced stage disease would benefit from a novel therapy preventing or reversing these changes. To this end, we have investigated the unique WR domain of the transcription factor TWIST1, which has been shown to play a role in driving metastasis and drug resistance. METHODS: In this study, we identified evolutionarily well-conserved residues within the TWIST1 WR domain and used alanine substitution to determine their role in WR domain-mediated protein binding. Co-immunoprecipitation was used to assay binding affinity between TWIST1 and the NFκB subunit p65 (RELA). Biological activity of this complex was assayed using a dual luciferase assay system in which firefly luciferase was driven by the interleukin-8 (IL-8) promoter, which is upregulated by the TWIST1-RELA complex. Finally, in order to inhibit the TWIST1-RELA interaction, we created a fusion protein comprising GFP and the WR domain. Cell fractionation and proteasome inhibition experiments were utilized to elucidate the mechanism of action of the GFP-WR fusion. RESULTS: We found that the central residues of the WR domain (W190, R191, E193) were important for TWIST1 binding to RELA, and for increased activation of the IL-8 promoter. We also found that the C-terminal 245 residues of RELA are important for TWIST1 binding and IL-8 promoter activation. Finally, we found the GFP-WR fusion protein antagonized TWIST1-RELA binding and downstream signaling. Co-expression of GFP-WR with TWIST1 and RELA led to proteasomal degradation of TWIST1, which could be inhibited by MG132 treatment. CONCLUSIONS: These data provide evidence that mutation or inhibition of the WR domain reduces TWIST1 activity, and may represent a potential therapeutic modality.


Interleukin-8/genetics , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Transcription Factor RelA/chemistry , Transcription Factor RelA/metabolism , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/metabolism , Binding Sites , HEK293 Cells , Humans , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Protein Domains , Transcriptional Activation , Twist-Related Protein 1/genetics
11.
Nucleic Acids Res ; 44(11): 5470-89, 2016 06 20.
Article En | MEDLINE | ID: mdl-27151200

The TWIST1 bHLH transcription factor controls embryonic development and cancer processes. Although molecular and genetic analyses have provided a wealth of data on the role of bHLH transcription factors, very little is known on the molecular mechanisms underlying their binding affinity to the E-box sequence of the promoter. Here, we used an in silico model of the TWIST1/E12 (TE) heterocomplex and performed molecular dynamics (MD) simulations of its binding to specific (TE-box) and modified E-box sequences. We focused on (i) active E-box and inactive E-box sequences, on (ii) modified active E-box sequences, as well as on (iii) two box sequences with modified adjacent bases the AT- and TA-boxes. Our in silico models were supported by functional in vitro binding assays. This exploration highlighted the predominant role of protein side-chain residues, close to the heart of the complex, at anchoring the dimer to DNA sequences, and unveiled a shift towards adjacent ((-1) and (-1*)) bases and conserved bases of modified E-box sequences. In conclusion, our study provides proof of the predictive value of these MD simulations, which may contribute to the characterization of specific inhibitors by docking approaches, and their use in pharmacological therapies by blocking the tumoral TWIST1/E12 function in cancers.


E-Box Elements , Models, Molecular , Transcription Factor 3/chemistry , Twist-Related Protein 1/chemistry , Base Sequence , Binding Sites , Cell Line , Humans , Hydrogen Bonding , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Binding , Transcription Factor 3/metabolism , Twist-Related Protein 1/metabolism
12.
Am J Hum Genet ; 97(1): 99-110, 2015 Jul 02.
Article En | MEDLINE | ID: mdl-26119818

Ablepharon macrostomia syndrome (AMS) and Barber-Say syndrome (BSS) are rare congenital ectodermal dysplasias characterized by similar clinical features. To establish the genetic basis of AMS and BSS, we performed extensive clinical phenotyping, whole exome and candidate gene sequencing, and functional validations. We identified a recurrent de novo mutation in TWIST2 in seven independent AMS-affected families, as well as another recurrent de novo mutation affecting the same amino acid in ten independent BSS-affected families. Moreover, a genotype-phenotype correlation was observed, because the two syndromes differed based solely upon the nature of the substituting amino acid: a lysine at TWIST2 residue 75 resulted in AMS, whereas a glutamine or alanine yielded BSS. TWIST2 encodes a basic helix-loop-helix transcription factor that regulates the development of mesenchymal tissues. All identified mutations fell in the basic domain of TWIST2 and altered the DNA-binding pattern of Flag-TWIST2 in HeLa cells. Comparison of wild-type and mutant TWIST2 expressed in zebrafish identified abnormal developmental phenotypes and widespread transcriptome changes. Our results suggest that autosomal-dominant TWIST2 mutations cause AMS or BSS by inducing protean effects on the transcription factor's DNA binding.


Abnormalities, Multiple/genetics , Eye Abnormalities/genetics , Eyelid Diseases/genetics , Hirsutism/genetics , Hypertelorism/genetics , Hypertrichosis/genetics , Macrostomia/genetics , Models, Molecular , Phenotype , Repressor Proteins/genetics , Skin Abnormalities/genetics , Twist-Related Protein 1/genetics , Abnormalities, Multiple/pathology , Amino Acid Sequence , Animals , Base Sequence , Chromatin Immunoprecipitation , Exome/genetics , Eye Abnormalities/pathology , Eyelid Diseases/pathology , HeLa Cells , Hirsutism/pathology , Humans , Hypertelorism/pathology , Hypertrichosis/pathology , Macrostomia/pathology , Microscopy, Electron , Molecular Sequence Data , Mutation, Missense/genetics , Protein Conformation , Repressor Proteins/chemistry , Sequence Analysis, DNA , Skin Abnormalities/pathology , Twist-Related Protein 1/chemistry , Zebrafish
13.
Genes Dev ; 29(6): 603-16, 2015 Mar 15.
Article En | MEDLINE | ID: mdl-25762439

Basic helix-loop-helix (bHLH) transcription factors recognize the canonical E-box (CANNTG) to regulate gene transcription; however, given the prevalence of E-boxes in a genome, it has been puzzling how individual bHLH proteins selectively recognize E-box sequences on their targets. TWIST is a bHLH transcription factor that promotes epithelial-mesenchymal transition (EMT) during development and tumor metastasis. High-resolution mapping of TWIST occupancy in human and Drosophila genomes reveals that TWIST, but not other bHLH proteins, recognizes a unique double E-box motif with two E-boxes spaced preferentially by 5 nucleotides. Using molecular modeling and binding kinetic analyses, we found that the strict spatial configuration in the double E-box motif aligns two TWIST-E47 dimers on the same face of DNA, thus providing a high-affinity site for a highly stable intramolecular tetramer. Biochemical analyses showed that the WR domain of TWIST dimerizes to mediate tetramer formation, which is functionally required for TWIST-induced EMT. These results uncover a novel mechanism for a bHLH transcription factor to recognize a unique spatial configuration of E-boxes to achieve target specificity. The WR-WR domain interaction uncovered here sets an example of target gene specificity of a bHLH protein being controlled allosterically by a domain outside of the bHLH region.


Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Models, Molecular , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/metabolism , Amino Acid Sequence , Animals , Biological Evolution , Conserved Sequence , Drosophila/chemistry , Drosophila/metabolism , Gene Expression Regulation , Humans , Protein Binding , Protein Stability , Protein Structure, Tertiary , Substrate Specificity
14.
Neoplasia ; 17(1): 16-31, 2015 Jan.
Article En | MEDLINE | ID: mdl-25622896

The TWIST1 gene has diverse roles in development and pathologic diseases such as cancer. TWIST1 is a dimeric basic helix-loop-helix (bHLH) transcription factor existing as TWIST1-TWIST1 or TWIST1-E12/47. TWIST1 partner choice and DNA binding can be influenced during development by phosphorylation of Thr125 and Ser127 of the Thr-Gln-Ser (TQS) motif within the bHLH of TWIST1. The significance of these TWIST1 phosphorylation sites for metastasis is unknown. We created stable isogenic prostate cancer cell lines overexpressing TWIST1 wild-type, phospho-mutants, and tethered versions. We assessed these isogenic lines using assays that mimic stages of cancer metastasis. In vitro assays suggested the phospho-mimetic Twist1-DQD mutation could confer cellular properties associated with pro-metastatic behavior. The hypo-phosphorylation mimic Twist1-AQA mutation displayed reduced pro-metastatic activity compared to wild-type TWIST1 in vitro, suggesting that phosphorylation of the TWIST1 TQS motif was necessary for pro-metastatic functions. In vivo analysis demonstrates that the Twist1-AQA mutation exhibits reduced capacity to contribute to metastasis, whereas the expression of the Twist1-DQD mutation exhibits proficient metastatic potential. Tethered TWIST1-E12 heterodimers phenocopied the Twist1-DQD mutation for many in vitro assays, suggesting that TWIST1 phosphorylation may result in heterodimerization in prostate cancer cells. Lastly, the dual phosphatidylinositide 3-kinase (PI3K)-mammalian target of rapamycin (mTOR) inhibitor BEZ235 strongly attenuated TWIST1-induced migration that was dependent on the TQS motif. TWIST1 TQS phosphorylation state determines the intensity of TWIST1-induced pro-metastatic ability in prostate cancer cells, which may be partly explained mechanistically by TWIST1 dimeric partner choice.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Nuclear Proteins/metabolism , Prostatic Neoplasms/metabolism , Protein Interaction Domains and Motifs , Twist-Related Protein 1/metabolism , Amino Acid Motifs , Animals , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cluster Analysis , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Gene Expression Profiling , Heterografts , Humans , Male , Mutation , Neoplasm Metastasis , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Multimerization , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , TOR Serine-Threonine Kinases/metabolism , Transcriptome , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/genetics
15.
Res Vet Sci ; 97(3): 521-6, 2014 Dec.
Article En | MEDLINE | ID: mdl-25278141

Epithelial-mesenchymal transition (EMT) is a crucial step in tumour progression. However, the molecular mechanisms underlying EMT in canine tumours remain to be elucidated. In this study, the similarity or difference in the molecular mechanism of EMT in canine cells was evaluated and compared with that reported in human and mouse cells. We used eight cell lines derived from canine mammary cancers. Stimulation with hepatocyte growth factor (HGF) increased cell motility and changed EMT-related markers towards mesenchyme in CHMm cell line. These changes were accompanied by an increase in Twist expression and did not occur in CHMm transfected with Twist siRNA, indicating that Twist plays a key role in this phenomenon in CHMm. However, the down-regulation of E-cadherin was not observed by HGF stimulation. Further studies are required to elucidate the difference between human and canine Twist.


Dog Diseases/genetics , Epithelial-Mesenchymal Transition , Gene Expression Regulation , Mammary Neoplasms, Animal/genetics , Twist-Related Protein 1/genetics , Up-Regulation , Amino Acid Sequence , Animals , Biomarkers/metabolism , Cell Line, Tumor , Cell Movement , Dog Diseases/physiopathology , Dogs , Female , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Mammary Neoplasms, Animal/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Transfection , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/metabolism
16.
J Biomol Struct Dyn ; 32(2): 226-41, 2014.
Article En | MEDLINE | ID: mdl-23527594

The basic helix-loop-helix (bHLH) transcription factor TWIST1 is essential to embryonic development, and hijacking of its function contributes to the development of numerous cancer types. It forms either a homodimer or a heterodimeric complex with an E2A or HAND partner. These functionally distinct complexes display sometimes antagonistic functions during development, so that alterations in the balance between them lead to pronounced morphological alterations, as observed in mice and in Saethre-Chotzen syndrome patients. We, here, describe the structures of TWIST1 bHLH-DNA complexes produced in silico through molecular dynamics simulations. We highlight the determinant role of the interhelical loops in maintaining the TWIST1-DNA complex structures and provide a structural explanation for the loss of function associated with several TWIST1 mutations/insertions observed in Saethre-Chotzen syndrome patients. An animated interactive 3D complement (I3DC) is available in Proteopedia at http://proteopedia.org/w/Journal:JBSD:27.


Helix-Loop-Helix Motifs , Nuclear Proteins/chemistry , Twist-Related Protein 1/chemistry , Acrocephalosyndactylia/genetics , Amino Acid Sequence , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/ultrastructure , Crystallography, X-Ray , DNA/chemistry , Humans , Mice , Molecular Dynamics Simulation , Mutation , MyoD Protein/chemistry , MyoD Protein/ultrastructure , Nuclear Proteins/genetics , Protein Binding/genetics , Protein Multimerization , Sequence Alignment , Transcription Factor 3/chemistry , Transcription Factor 3/ultrastructure , Twist-Related Protein 1/genetics
17.
Dev Genes Evol ; 223(5): 319-28, 2013 Sep.
Article En | MEDLINE | ID: mdl-23817621

The basic helix-loop-helix transcription factor twist plays a key role during mesoderm development in Bilateria. In this study, we identified a twist ortholog in the polychaete annelid Platynereis dumerilii and analyze its expression during larval development, postlarval growth up to the adult stage, and caudal regeneration after amputation of posterior segments. At late larval stages, Pdu-twist is expressed in the mesodermal anlagen and in developing muscles. During adulthood and caudal regeneration, Pdu-twist is expressed in the posterior growth zone, in mesodermal cells within the newly forming segments and budding parapodia. Our results indicate that Pdu-twist is involved in mesoderm formation during larval development, posterior growth, and caudal regeneration.


Polychaeta/embryology , Polychaeta/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Mesoderm/metabolism , Molecular Sequence Data , Phylogeny , Twist-Related Protein 1/chemistry
18.
Nat Commun ; 4: 1542, 2013.
Article En | MEDLINE | ID: mdl-23443570

A subset of transcription factors classified as neural crest 'specifiers' are also core epithelial-mesenchymal transition regulatory factors, both in the neural crest and in tumour progression. The bHLH factor Twist is among the least well studied of these factors. Here we demonstrate that Twist is required for cranial neural crest formation and fate determination in Xenopus. We further show that Twist function in the neural crest is dependent upon its carboxy-terminal WR domain. The WR domain mediates physical interactions between Twist and other core epithelial-mesenchymal transition factors, including Snail1 and Snail2, which are essential for proper function. Interaction with Snail1/2, and Twist function more generally, is regulated by GSK-3-ß-mediated phosphorylation of conserved sites in the WR domain. Together, these findings elucidate a mechanism for coordinated control of a group of structurally diverse factors that function as a regulatory unit in both developmental and pathological epithelial-mesenchymal transitions.


Epithelial-Mesenchymal Transition , Glycogen Synthase Kinase 3/metabolism , Transcription Factors/metabolism , Twist-Related Protein 1/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Amino Acid Sequence , Animals , Blotting, Western , Body Patterning/genetics , Cell Movement , Gene Expression Regulation, Developmental , Immunoprecipitation , In Situ Hybridization , Molecular Sequence Data , Mutation/genetics , Neural Crest/cytology , Neural Crest/enzymology , Neural Crest/growth & development , Phosphorylation , Protein Binding , Protein Stability , Protein Structure, Tertiary , Snail Family Transcription Factors , Substrate Specificity , Twist-Related Protein 1/chemistry , Xenopus Proteins/chemistry , Xenopus laevis/genetics
19.
BMC Bioinformatics ; 13: 184, 2012 Jul 28.
Article En | MEDLINE | ID: mdl-22839202

BACKGROUND: Human TWIST1 is a highly conserved member of the regulatory basic helix-loop-helix (bHLH) transcription factors. TWIST1 forms homo- or heterodimers with E-box proteins, such as E2A (isoforms E12 and E47), MYOD and HAND2. Haploinsufficiency germ-line mutations of the twist1 gene in humans are the main cause of Saethre-Chotzen syndrome (SCS), which is characterized by limb abnormalities and premature fusion of cranial sutures. Because of the importance of TWIST1 in the regulation of embryonic development and its relationship with SCS, along with the lack of an experimentally solved 3D structure, we performed comparative modeling for the TWIST1 bHLH region arranged into wild-type homodimers and heterodimers with E47. In addition, three mutations that promote DNA binding failure (R118C, S144R and K145E) were studied on the TWIST1 monomer. We also explored the behavior of the mutant forms in aqueous solution using molecular dynamics (MD) simulations, focusing on the structural changes of the wild-type versus mutant dimers. RESULTS: The solvent-accessible surface area of the homodimers was smaller on wild-type dimers, which indicates that the cleft between the monomers remained more open on the mutant homodimers. RMSD and RMSF analyses indicated that mutated dimers presented values that were higher than those for the wild-type dimers. For a more careful investigation, the monomer was subdivided into four regions: basic, helix I, loop and helix II. The basic domain presented a higher flexibility in all of the parameters that were analyzed, and the mutant dimer basic domains presented values that were higher than the wild-type dimers. The essential dynamic analysis also indicated a higher collective motion for the basic domain. CONCLUSIONS: Our results suggest the mutations studied turned the dimers into more unstable structures with a wider cleft, which may be a reason for the loss of DNA binding capacity observed for in vitro circumstances.


Computer Simulation , Helix-Loop-Helix Motifs , Models, Chemical , Nuclear Proteins/chemistry , Twist-Related Protein 1/chemistry , Acrocephalosyndactylia/genetics , Acrocephalosyndactylia/metabolism , Amino Acid Sequence , Amino Acid Substitution , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Protein Multimerization , Protein Structure, Secondary , Twist-Related Protein 1/genetics
20.
BMC Cancer ; 11: 350, 2011 Aug 11.
Article En | MEDLINE | ID: mdl-21834956

BACKGROUND: Epithelial-mesenchymal transition (EMT) is a crucial process in tumorigenesis since tumor cells attain fibroblast-like features enabling them to invade to surrounding tissue. Two transcription factors, TWIST and SNAI1, are fundamental in regulating EMT. METHODS: Immunohistochemistry was used to study the expression of TWIST and SNAI1 in 109 pharyngeal squamous cell carcinomas. RESULTS: Tumors with intense stromal staining of TWIST relapsed more frequently (p = 0.04). Tumors with both positive TWIST and SNAI1 immunoreactivity in the stroma were at least Stage II (p = 0.05) and located more often in hypopharynx (p = 0.035). Tumors with negative immunostaining of TWIST and SNAI1 in the stromal compartment were smaller (T1-2) (p = 0.008), less advanced (SI-II) (p = 0.031) and located more often in the oropharynx (p = 0.007). Patients with negative SNAI1 and TWIST immunostaining in tumor stroma had a better 5-year disease-specific and overall survival (p = 0.037 and p = 0.014 respectively). CONCLUSION: TWIST and SNAI1 expression in stromal cells is associated with clinical and histopathological characteristics that indicate progressive disease. Negative expression of these EMT-promoting transcription factors predicts a better outcome.


Carcinoma, Squamous Cell/metabolism , Nuclear Proteins/biosynthesis , Pharyngeal Neoplasms/metabolism , Stromal Cells/metabolism , Transcription Factors/biosynthesis , Twist-Related Protein 1/biosynthesis , Adult , Aged , Aged, 80 and over , Analysis of Variance , Carcinoma, Squamous Cell/pathology , Chi-Square Distribution , Disease Progression , Epithelial-Mesenchymal Transition , Female , Finland , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Longitudinal Studies , Male , Middle Aged , Nuclear Proteins/chemistry , Pharyngeal Neoplasms/pathology , Prognosis , Snail Family Transcription Factors , Stromal Cells/pathology , Transcription Factors/chemistry , Twist-Related Protein 1/chemistry
...