Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38966985

RESUMO

The TOPOVIL complex catalyzes the formation of DNA double strand breaks (DSB) that initiate meiotic homologous recombination, an essential step for chromosome segregation and genetic diversity during gamete production. TOPOVIL is composed of two subunits (SPO11 and TOPOVIBL) and is evolutionarily related to the archaeal TopoVI topoisomerase complex. SPO11 is the TopoVIA subunit orthologue and carries the DSB formation catalytic activity. TOPOVIBL shares homology with the TopoVIB ATPase subunit. TOPOVIBL is essential for meiotic DSB formation, but its molecular function remains elusive, partly due to the lack of biochemical studies. Here, we purified TOPOVIBLΔC25 and characterized its structure and mode of action in vitro. Our structural analysis revealed that TOPOVIBLΔC25 adopts a dynamic conformation in solution and our biochemical study showed that the protein remains monomeric upon incubation with ATP, which correlates with the absence of ATP binding. Moreover, TOPOVIBLΔC25 interacted with DNA, with a preference for some geometries, suggesting that TOPOVIBL senses specific DNA architectures. Altogether, our study identified specific TOPOVIBL features that might help to explain how TOPOVIL function evolved toward a DSB formation activity in meiosis.

2.
Subcell Biochem ; 70: 37-54, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24962880

RESUMO

In the form of heterodimers with retinoid X receptors (RXRs), retinoic acid receptors (RARs) are master regulators of gene expression in humans and important drug targets. They act as ligand-dependent transcription factors that regulate a large variety of gene networks controlling cell growth, differentiation, survival and death. The biological functions of RARs rely on a dynamic series of coregulator exchanges controlled by ligand binding. Unliganded RARs exert a repressor activity by interacting with transcriptional corepressors which themselves serve as docking platforms for the recruitment of histone deacetylases that impose a higher order structure on chromatin which is not permissive to gene transcription. Upon ligand binding, the receptor undergoes conformational changes inducing corepressor release and the recruitment of coactivators with histone acetylase activities allowing chromatin decompaction and gene transcription. In the following, we review the structural determinants of the interaction between RAR and either type of coregulators both at the level of the individual receptor and in the context of the RAR-RXR heterodimers. We also discuss the molecular details of the fine tuning of these associations by the various pharmacological classes of ligands.


Assuntos
Cromatina/química , Regulação da Expressão Gênica , Receptores do Ácido Retinoico/química , Receptores X de Retinoides/química , Cromatina/metabolismo , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Humanos , Ligantes , Modelos Moleculares , Ligação Proteica , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Multimerização Proteica , Estrutura Terciária de Proteína , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Elementos de Resposta , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo , Transdução de Sinais
3.
Proc Natl Acad Sci U S A ; 109(37): 14930-5, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22927406

RESUMO

Bisphenol A (BPA) is an industrial compound and a well known endocrine-disrupting chemical with estrogenic activity. The widespread exposure of individuals to BPA is suspected to affect a variety of physiological functions, including reproduction, development, and metabolism. Here we report that the mechanisms by which BPA and two congeners, bisphenol AF and bisphenol C (BPC), bind to and activate estrogen receptors (ER) α and ß differ from that used by 17ß-estradiol. We show that bisphenols act as partial agonists of ERs by activating the N-terminal activation function 1 regardless of their effect on the C-terminal activation function 2, which ranges from weak agonism (with BPA) to antagonism (with BPC). Crystallographic analysis of the interaction between bisphenols and ERs reveals two discrete binding modes, reflecting the different activities of compounds on ERs. BPA and 17ß-estradiol bind to ERs in a similar fashion, whereas, with a phenol ring pointing toward the activation helix H12, the orientation of BPC accounts for the marked antagonist character of this compound. Based on structural data, we developed a protocol for in silico evaluation of the interaction between bisphenols and ERs or other members of the nuclear hormone receptor family, such as estrogen-related receptor γ and androgen receptor, which are two known main targets of bisphenols. Overall, this study provides a wealth of tools and information that could be used for the development of BPA substitutes devoid of nuclear hormone receptor-mediated activity and more generally for environmental risk assessment.


Assuntos
Disruptores Endócrinos/metabolismo , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Modelos Moleculares , Fenóis/metabolismo , Animais , Compostos Benzidrílicos , Linhagem Celular , Cromatografia em Gel , Cristalografia , Relação Dose-Resposta a Droga , Disruptores Endócrinos/química , Estradiol/metabolismo , Receptor alfa de Estrogênio/isolamento & purificação , Receptor beta de Estrogênio/isolamento & purificação , Polarização de Fluorescência , Humanos , Luciferases , Oncorhynchus mykiss , Fenóis/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
J Mol Endocrinol ; 69(3): 377-390, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35900852

RESUMO

Retinoid X receptors (RXRα, ß, and γ) are essential members of the nuclear receptor (NR) superfamily of ligand-dependent transcriptional regulators that bind DNA response elements and control the expression of large gene networks. As obligate heterodimerization partners of many NRs, RXRs are involved in a variety of pathophysiological processes. However, despite this central role in NR signaling, there is still no consensus regarding the precise biological functions of RXRs and the putative role of the endogenous ligands (rexinoids) previously proposed for these receptors. Based on available crystal structures, we introduced a series of amino acid substitutions into the ligand-binding pocket of all three RXR subtypes in order to alter their binding properties. Subsequent characterization using a battery of cell-based and in vitro assays led to the identification of a double mutation abolishing the binding of any ligand while keeping the other receptor functions intact and a triple mutation that selectively impairs interaction with natural rexinoids but not with some synthetic ligands. We also report crystal structures that help understand the specific ligand-binding capabilities of both variants. These RXR variants, either fully disabled for ligand binding or retaining the property of being activated by synthetic compounds, represent unique tools that could be used in future studies to probe the presence of active endogenous rexinoids in tissues/organs and to investigate their role in vivo. Last, we provide data suggesting a possible involvement of fatty acids in the weak interaction of RXRs with corepressors.


Assuntos
Receptores Citoplasmáticos e Nucleares , Transdução de Sinais , Regulação da Expressão Gênica , Ligantes , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores X de Retinoides/química , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo
5.
Acta Crystallogr D Biol Crystallogr ; 67(Pt 9): 747-55, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21904027

RESUMO

X-ray crystallography is now a recognized technique for ligand screening, especially for fragment-based drug design. However, protein crystal handling is still tedious and limits further automation. An alternative method for the solution of crystal structures of proteins in complex with small ligands is proposed. Crystallization drops are directly exposed to an X-ray beam after cocrystallization or soaking with the desired ligands. The use of dedicated plates in connection with an optimal parametrization of the G-rob robot allows efficient data collection. Three proteins currently under study in our laboratory for ligand screening by X-ray crystallography were used as validation test cases. The protein crystals belonged to different space groups, including a challenging monoclinic case. The resulting diffraction data can lead to clear ligand recognition, including indication of alternating conformations. These results demonstrate a possible method for automation of ligand screening by X-ray crystallography.


Assuntos
Cristalografia por Raios X/métodos , Proteínas/química , Difração de Raios X/métodos , Desenho de Fármacos
6.
EMBO Rep ; 10(4): 367-73, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19270714

RESUMO

The nuclear receptor retinoid X receptor-alpha (RXR-alpha)-peroxisome proliferator-activated receptor-gamma (PPAR-gamma) heterodimer was recently reported to have a crucial function in mediating the deleterious effects of organotin compounds, which are ubiquitous environmental contaminants. However, because organotins are unrelated to known RXR-alpha and PPAR-gamma ligands, the mechanism by which these compounds bind to and activate the RXR-alpha-PPAR-gamma heterodimer at nanomolar concentrations has remained elusive. Here, we show that tributyltin (TBT) activates all three RXR-PPAR-alpha, -gamma, -delta heterodimers, primarily through its interaction with RXR. In addition, the 1.9 A resolution structure of the RXR-alpha ligand-binding domain in complex with TBT shows a covalent bond between the tin atom and residue Cys 432 of helix H11. This interaction largely accounts for the high binding affinity of TBT, which only partly occupies the RXR-alpha ligand-binding pocket. Our data allow an understanding of the binding and activation properties of the various organotins and suggest a mechanism by which these tin compounds could affect other nuclear receptor signalling pathways.


Assuntos
Disruptores Endócrinos/farmacologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Multimerização Proteica/efeitos dos fármacos , Receptores X de Retinoides/metabolismo , Compostos de Trialquitina/farmacologia , Linhagem Celular , Cromatografia Líquida , Cristalografia por Raios X , Disruptores Endócrinos/química , Polarização de Fluorescência , Humanos , Espectrometria de Massas , Modelos Biológicos , Estrutura Molecular , Receptores Ativados por Proliferador de Peroxissomo/química , Estrutura Secundária de Proteína , Receptores X de Retinoides/química , Compostos de Trialquitina/química
7.
Cell Mol Life Sci ; 67(8): 1219-37, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20063036

RESUMO

Endocrine-disrupting chemicals (EDCs) represent a broad class of exogenous substances that cause adverse effects in the endocrine system by interfering with hormone biosynthesis, metabolism, or action. The molecular mechanisms of EDCs involve different pathways including interactions with nuclear hormone receptors (NHRs) which are primary targets of a large variety of environmental contaminants. Here, based on the crystal structures currently available in the Protein Data Bank, we review recent studies showing the many ways in which EDCs interact with NHRs and impact their signaling pathways. Like the estrogenic chemical diethylstilbestrol, some EDCs mimic the natural hormones through conserved protein-ligand contacts, while others, such as organotins, employ radically different binding mechanisms. Such structure-based knowledge, in addition to providing a better understanding of EDC activities, can be used to predict the endocrine-disrupting potential of environmental pollutants and may have applications in drug discovery.


Assuntos
Disruptores Endócrinos/farmacologia , Sistema Endócrino/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Humanos
8.
J Mol Biol ; 433(9): 166899, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33647291

RESUMO

Retinoic acid receptors (RARs) and retinoid X receptors (RXRs) form heterodimers that activate target gene transcription by recruiting co-activator complexes in response to ligand binding. The nuclear receptor (NR) co-activator TIF2 mediates this recruitment by interacting with the ligand-binding domain (LBD) of NRs trough the nuclear receptor interaction domain (TIF2NRID) containing three highly conserved α-helical LxxLL motifs (NR-boxes). The precise binding mode of this domain to RXR/RAR is not clear due to the disordered nature of TIF2. Here we present the structural characterization of TIF2NRID by integrating several experimental (NMR, SAXS, Far-UV CD, SEC-MALS) and computational data. Collectively, the data are in agreement with a largely disordered protein with partially structured regions, including the NR-boxes and their flanking regions, which are evolutionary conserved. NMR and X-ray crystallographic data on TIF2NRID in complex with RXR/RAR reveal a multisite binding of the three NR-boxes as well as an active role of their flanking regions in the interaction.


Assuntos
Coativador 2 de Receptor Nuclear/química , Coativador 2 de Receptor Nuclear/metabolismo , Receptores do Ácido Retinoico/química , Receptores do Ácido Retinoico/metabolismo , Receptores X de Retinoides/química , Receptores X de Retinoides/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cristalografia por Raios X , Polarização de Fluorescência , Humanos , Proteínas Intrinsicamente Desordenadas/química , Proteínas Intrinsicamente Desordenadas/metabolismo , Ligantes , Camundongos , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Multimerização Proteica , Estrutura Secundária de Proteína
9.
Methods Enzymol ; 637: 175-207, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32359645

RESUMO

The three retinoic acid receptor subtypes (RARα, RARß and RARγ) act as ligand-inducible transcription factors binding to DNA regulatory elements in the promoter regions of target genes by forming heterodimers with the retinoid X receptors (RXRα, RXRß and RXRγ). They act as ligand-dependent transcription factors that regulate a large variety of genes involved in cell growth, differentiation, survival and death. The (patho)physiological functions of RAR-RXR heterodimers rely on a dynamic sequence of protein-protein interactions, many of which being modulated by natural (retinoic acid) or synthetic ligands. Direct protein-protein interactions include heterodimerization between RARs and RXRs, recruitment (and release) of transcriptional coactivators and corepressors, cross-talk with other transcription factors, including nuclear receptors, or transient association with many enzymes involved in post-translational modifications to cite the most prominent ones. This chapter describes structural, biochemical, biophysical and cell-based assays to monitor protein-protein interactions relevant to the retinoic acid signaling pathways with a focus on those for which a structural description has been provided.


Assuntos
Transdução de Sinais , Tretinoína , Regulação da Expressão Gênica , Ligantes , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo
10.
Prog Mol Biol Transl Sci ; 174: 105-155, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32828464

RESUMO

G protein-coupled receptors (GPCRs) and Nuclear Receptors (NRs) are two signaling machineries that are involved in major physiological processes and, as a consequence, in a substantial number of diseases. Therefore, they actually represent two major targets for drugs with potential applications in almost all public health issues. Full exploitation of these targets for therapeutic purposes nevertheless requires opening original avenues in drug design, and this in turn implies a better understanding of the molecular mechanisms underlying their functioning. However, full comprehension of how these complex systems function and how they are deregulated in a physiopathological context is obscured by the fact that these proteins include a substantial number of disordered regions that are central to their mechanism of action but whose structural and functional properties are still largely unexplored. In this chapter, we describe how these intrinsically disordered regions (IDR) or proteins (IDP) intervene, control and finely modulate the thermodynamics of complexes involved in GPCR and NR regulation, which in turn triggers a multitude of cascade of events that are exquisitely orchestrated to ultimately control the biological output.


Assuntos
Proteínas Intrinsicamente Desordenadas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Humanos , Proteínas Intrinsicamente Desordenadas/química , Modelos Moleculares , Processamento de Proteína Pós-Traducional , Receptores Acoplados a Proteínas G/química
11.
Front Endocrinol (Lausanne) ; 11: 561256, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329381

RESUMO

The nuclear receptor PPARγ is essential to maintain whole-body glucose homeostasis and insulin sensitivity, acting as a master regulator of adipogenesis, lipid, and glucose metabolism. Its activation through natural or synthetic ligands induces the recruitment of coactivators, leading to transcription of target genes such as cytokines and hormones. More recently, post translational modifications, such as PPARγ phosphorylation at Ser273 by CDK5 in adipose tissue, have been linked to insulin resistance trough the dysregulation of expression of a specific subset of genes. Here, we investigate how this phosphorylation may disturb the interaction between PPARγ and some coregulator proteins as a new mechanism that may leads to insulin resistance. Through cellular and in vitro assays, we show that PPARγ phosphorylation inhibition increased the activation of the receptor, therefore the increased recruitment of PGC1-α and TIF2 coactivators, whilst decreases the interaction with SMRT and NCoR corepressors. Moreover, our results show a shift in the coregulators interaction domains preferences, suggesting additional interaction interfaces formed between the phosphorylated PPARγ and some coregulator proteins. Also, we observed that the CDK5 presence disturb the PPARγ-coregulator's synergy, decreasing interaction with PGC1-α, TIF2, and NCoR, but increasing coupling of SMRT. Finally, we conclude that the insulin resistance provoked by PPARγ phosphorylation is linked to a differential coregulators recruitment, which may promote dysregulation in gene expression.


Assuntos
Resistência à Insulina/fisiologia , PPAR gama/metabolismo , Serina/metabolismo , Células 3T3 , Adipócitos/metabolismo , Animais , Células COS , Chlorocebus aethiops , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Células HEK293 , Humanos , Camundongos , PPAR gama/genética , Fosforilação/fisiologia , Serina/genética
12.
Thyroid ; 30(8): 1217-1221, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32204686

RESUMO

Resistance to thyroid hormone alpha (RTHα) is a rare and under-recognized genetic disease caused by mutations of THRA, the gene encoding thyroid hormone receptor α1 (TRα1). We report here two novel THRA missense mutations (M259T, T273A) in patients with RTHα. We combined biochemical and cellular assays with in silico modeling to assess the capacity of mutant TRα1 to bind triiodothyronine (T3), to heterodimerize with RXR, to interact with transcriptional coregulators, and to transduce a T3 transcriptional response. M259T, and to a lower extent T273A, reduces the affinity of TRα1 for T3. Their negative influence is only reverted by large excess of T3. The severity of the two novel RTHα cases originates from a reduction in the binding affinity of TRα1 mutants to T3 and thus correlates with the incapacity of corepressors to dissociate from TRα1 mutants in the presence of T3.


Assuntos
Mutação de Sentido Incorreto , Receptores alfa dos Hormônios Tireóideos/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Pré-Escolar , Simulação por Computador , Dimerização , Feminino , Heterozigoto , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Ligantes , Mutação , Fenótipo , Síndrome da Resistência aos Hormônios Tireóideos/sangue , Hormônios Tireóideos , Tiroxina/metabolismo , Ativação Transcricional , Transfecção , Tri-Iodotironina/metabolismo
13.
Cells ; 8(11)2019 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-31694317

RESUMO

The three subtypes (α, ß, and γ) of the retinoic acid receptor (RAR) are ligand-dependent transcription factors that mediate retinoic acid signaling by forming heterodimers with the retinoid X receptor (RXR). Heterodimers are functional units that bind ligands (retinoids), transcriptional co-regulators and DNA, to regulate gene networks controlling cell growth, differentiation, and death. Using biochemical, crystallographic, and cellular approaches, we have set out to explore the spectrum of possibilities to regulate RXR-RAR heterodimer-dependent transcription through various pharmacological classes of RAR- and RXR- specific ligands, alone or in combination. We reveal the molecular details by which these compounds direct specificity and functionality of RXR-RAR heterodimers. Among these ligands, we have reevaluated and improved the molecular and structural definition of compounds CD2665, Ro41-5253, LE135, or LG100754, highlighting novel functional features of these molecules. Our analysis reveals a model of RXR-RAR heterodimer action in which each subunit retains its intrinsic properties in terms of ligand and co-regulator binding. However, their interplay upon the combined action of RAR- and RXR-ligands allows for the fine tuning of heterodimer activity. It also stresses the importance of accurate ligand characterization to use synthetic selective retinoids appropriately and avoid data misinterpretations.


Assuntos
Receptores do Ácido Retinoico/metabolismo , Receptores X de Retinoides/metabolismo , Adamantano/análogos & derivados , Adamantano/farmacologia , Benzoatos/farmacologia , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Cromanos/farmacologia , Dibenzazepinas/farmacologia , Dimerização , Humanos , Ligantes , Células MCF-7 , Subunidades Proteicas/metabolismo , Retinoides/farmacologia , Tetra-Hidronaftalenos/farmacologia
14.
Structure ; 27(8): 1270-1285.e6, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31178221

RESUMO

In its unliganded form, the retinoic acid receptor (RAR) in heterodimer with the retinoid X receptor (RXR) exerts a strong repressive activity facilitated by the recruitment of transcriptional corepressors in the promoter region of target genes. By integrating complementary structural, biophysical, and computational information, we demonstrate that intrinsic disorder is a required feature for the precise regulation of RAR activity. We show that structural dynamics of RAR and RXR H12 regions is an essential mechanism for RAR regulation. Unexpectedly we found that, while mainly disordered, the corepressor N-CoR presents evolutionary conserved structured regions involved in transient intramolecular contacts. In the presence of RXR/RAR, N-CoR exploits its multivalency to form a cooperative multisite complex that displays equilibrium between different conformational states that can be tuned by cognate ligands and receptor mutations. This equilibrium is key to preserving the repressive basal state while allowing the conversion to a transcriptionally active form.


Assuntos
Correpressor 1 de Receptor Nuclear/genética , Receptor alfa de Ácido Retinoico/química , Receptor alfa de Ácido Retinoico/metabolismo , Receptores X de Retinoides/química , Receptores X de Retinoides/metabolismo , Animais , Células COS , Chlorocebus aethiops , Evolução Molecular , Regulação da Expressão Gênica , Humanos , Modelos Moleculares , Simulação de Dinâmica Molecular , Correpressor 1 de Receptor Nuclear/química , Correpressor 1 de Receptor Nuclear/metabolismo , Domínios Proteicos , Dobramento de Proteína , Multimerização Proteica , Estrutura Secundária de Proteína
15.
Thyroid ; 28(12): 1708-1722, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30235988

RESUMO

BACKGROUND: Thyroid hormone receptors (TRs) are tightly regulated by the corepressors nuclear receptor corepressor (NCoR) and silencing mediator of retinoic acid and thyroid hormone receptors. Three conserved corepressor/NR signature box motifs (CoRNR1-3) forming the nuclear receptor interaction domain have been identified in these corepressors. Whereas TRs regulate multiple normal physiological and developmental pathways, mutations in TRs can result in endocrine diseases and be associated with cancers due to impairment of corepressor release. Three mutants that are located in helix H11 of TRs are of special interest: TRα-M388I, a mutant associated with the development of renal clear cell carcinomas (RCCCs), and TRß-Δ430 and TRß-Δ432, two deletion mutants causing resistance to thyroid hormone syndrome. METHODS: Several cell-based and biophysical methods were used to measure the affinity between wild-type and mutant TRα and TRß and all the CoRNR motifs from corepressors to quantify the effects of different thyroid hormone analogues on these interactions. This study was coupled with the measurement of interactions between wild-type and mutant TRs in the context of a heterodimer with RXR to a NCoR fragment in the presence of the same ligands. Structural insights into the binding mode of corepressors to TRs were assessed in parallel by nuclear magnetic resonance spectroscopy. RESULTS: The study shows that TRs interact more avidly with the silencing mediator of retinoic acid and thyroid hormone receptors than with NCoR peptides, and that TRα binds most avidly to S-CoRNR3, whereas TRß binds preferentially to S-CoRNR2. In the studied TR mutants, a transfer of the CoRNR-specificity toward CoRNR1 was observed, coupled with a significant increase in the binding strength. In contrast to 3,5,3'-triiodothyronine (T3), the agonist TRIAC and the antagonist NH-3 were very efficient at dissociating the abnormally strong interactions between mutant TRßs and corepressors. A strong impairment of T3-binding for TRß mutants was shown compared to TRIAC and NH-3 and could explain the different efficiencies of the different ligands in releasing corepressors from the studied TRß mutants. Consequently, TRIAC was found to be more effective than T3 in facilitating coactivator recruitment and decreasing the dominant activity of TRß-Δ430. CONCLUSION: This study helps to clarify the specific interaction surfaces involved in the pathologic phenotype of TR mutants and demonstrates that TRIAC is a potential therapeutic agent for patients suffering from resistance to thyroid hormone syndromes.


Assuntos
Proteínas Correpressoras/metabolismo , Mutação , Receptores dos Hormônios Tireóideos/metabolismo , Síndrome da Resistência aos Hormônios Tireóideos/metabolismo , Hormônios Tireóideos/química , Anisotropia , Humanos , Cinética , Ligantes , Espectroscopia de Ressonância Magnética , Proteínas Nucleares/metabolismo , Peptídeos/química , Ligação Proteica , Receptores dos Hormônios Tireóideos/genética , Proteínas Repressoras/genética , Espectrometria de Fluorescência , Glândula Tireoide/metabolismo , Receptores alfa dos Hormônios Tireóideos/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Síndrome da Resistência aos Hormônios Tireóideos/genética
16.
Artigo em Inglês | MEDLINE | ID: mdl-29449830

RESUMO

Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of a nuclear receptor superfamily and acts as a ligand-dependent transcription factor, playing key roles in maintenance of adipose tissue and in regulation of glucose and lipid homeostasis. This receptor is the target of thiazolidinediones, a class of antidiabetic drugs, which improve insulin sensitization and regulate glycemia in type 2 diabetes. Despite the beneficial effects of drugs, such as rosiglitazone and pioglitazone, their use is associated with several side effects, including weight gain, heart failure, and liver disease, since these drugs induce full activation of the receptor. By contrast, a promising activation-independent mechanism that involves the inhibition of cyclin-dependent kinase 5 (CDK5)-mediated PPARγ phosphorylation has been related to the insulin-sensitizing effects induced by these drugs. Thus, we aimed to identify novel PPARγ ligands that do not possess agonist properties by conducting a mini-trial with 80 compounds using the sequential steps of thermal shift assay, 8-anilino-1-naphthalenesulfonic acid fluorescence quenching, and a cell-based transactivation assay. We identified two non-agonist PPARγ ligands, AM-879 and P11, and one partial-agonist, R32. Using fluorescence anisotropy, we show that AM-879 does not dissociate the NCOR corepressor in vitro, and it has only a small effect on TRAP coactivator recruitment. In cells, AM-879 could not induce adipocyte differentiation or positively regulate the expression of genes associated with adipogenesis. In addition, AM-879 inhibited CDK5-mediated phosphorylation of PPARγ in vitro. Taken together, these findings supported an interaction between AM-879 and PPARγ; this interaction was identified by the analysis of the crystal structure of the PPARγ:AM-879 complex and evidenced by AM-879's mechanism of action as a putative PPARγ non-agonist with antidiabetic properties. Moreover, we present an optimized assay pipeline capable of detecting ligands that physically bind to PPARγ but do not cause its activation as a new strategy to identify ligands for this nuclear receptor.

17.
Protein Sci ; 16(12): 2750-5, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18029424

RESUMO

Human KIN17 is a 45-kDa eukaryotic DNA- and RNA-binding protein that plays an important role in nuclear metabolism and in particular in the general response to genotoxics. Its amino acids sequence contains a zinc finger motif (residues 28-50) within a 30-kDa N-terminal region conserved from yeast to human, and a 15-kDa C-terminal tandem of SH3-like subdomains (residues 268-393) only found in higher eukaryotes. Here we report the solution structure of the region 51-160 of human KIN17. We show that this fragment folds into a three-alpha-helix bundle packed against a three-stranded beta-sheet. It belongs to the winged helix (WH) family. Structural comparison with analogous WH domains reveals that KIN17 WH module presents an additional and highly conserved 3(10)-helix. Moreover, KIN17 WH helix H3 is not positively charged as in classical DNA-binding WH domains. Thus, human KIN17 region 51-160 might rather be involved in protein-protein interaction through its conserved surface centered on the 3(10)-helix.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a RNA/química , Sequência de Aminoácidos , Animais , Proteínas de Ligação a DNA/isolamento & purificação , Vetores Genéticos , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/isolamento & purificação , Proteínas Recombinantes de Fusão , Alinhamento de Sequência , Fatores de Transcrição Winged-Helix/química , Dedos de Zinco
18.
J Mol Biol ; 364(4): 764-76, 2006 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-17045609

RESUMO

The human KIN17 protein is an essential nuclear protein conserved from yeast to human and expressed ubiquitously in mammals. Suppression of Rts2, the yeast equivalent of gene KIN17, renders the cells unviable, and silencing the human KIN17 gene slows cell growth dramatically. Moreover, the human gene KIN17 is up-regulated following exposure to ionizing radiations and UV light, depending on the integrity of the human global genome repair machinery. Its ectopic over-expression blocks S-phase progression by inhibiting DNA synthesis. The C-terminal region of human KIN17 is crucial for this anti-proliferation effect. Its high-resolution structure, presented here, reveals a tandem of SH3-like subdomains. This domain binds to ribonucleotide homopolymers with the same preferences as the whole protein. Analysis of its structure complexed with tungstate shows structural variability within the domain. The interaction with tungstate is mediated by several lysine residues located within a positively charged groove at the interface between the two subdomains. This groove could be the site of interaction with RNA, since mutagenesis of two of these highly conserved lysine residue weakens RNA binding.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a RNA/química , RNA/metabolismo , Sítios de Ligação , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Lisina , Mutagênicos/farmacologia , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas de Ligação a RNA/metabolismo , Compostos de Tungstênio/química , Domínios de Homologia de src
19.
Artigo em Inglês | MEDLINE | ID: mdl-16511313

RESUMO

Here, the crystallization and initial phasing of the C-terminal domain of human KIN17, a 45 kDa protein mainly expressed in response to ionizing radiation and overexpressed in certain tumour cell lines, are reported. Crystals diffracting to 1.4 A resolution were obtained from 10% ethylene glycol, 27% PEG 6000, 500 mM LiCl and 100 mM sodium acetate pH 6.3 in space group P2(1)2(1)2(1), with unit-cell parameters a = 45.75, b = 46.31, c = 60.80 A and one molecule in the asymmetric unit. Since this domain has a basic pI, heavy-atom derivatives were obtained by soaking the crystals with negatively charged ions such as tungstate and iodine. The replacement of LiCl by KI in the cryosolution allowed the determination of phases from iodide ions to give an interpretable electron-density map.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas Nucleares/química , Cloretos/química , Cristalização/métodos , Cristalografia por Raios X/métodos , Humanos , Iodetos/química , Fosfatos/química , Estrutura Terciária de Proteína/efeitos dos fármacos , Proteínas de Ligação a RNA , Compostos de Tungstênio/química
20.
PLoS One ; 11(11): e0165139, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27812132

RESUMO

Mixed Lineage Leukemia 5 (MLL5) plays a key role in hematopoiesis, spermatogenesis and cell cycle progression. Chromatin binding is ensured by its plant homeodomain (PHD) through a direct interaction with the N-terminus of histone H3 (H3). In addition, MLL5 contains a Su(var)3-9, Enhancer of zeste, Trithorax (SET) domain, a protein module that usually displays histone lysine methyltransferase activity. We report here the crystal structure of the unliganded SET domain of human MLL5 at 2.1 Å resolution. Although it shows most of the canonical features of other SET domains, both the lack of key residues and the presence in the SET-I subdomain of an unusually large loop preclude the interaction of MLL5 SET with its cofactor and substrate. Accordingly, we show that MLL5 is devoid of any in vitro methyltransferase activity on full-length histones and histone H3 peptides. Hence, the three dimensional structure of MLL5 SET domain unveils the structural basis for its lack of methyltransferase activity and suggests a new regulatory mechanism.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Sequência de Aminoácidos , Biocatálise , Cristalografia por Raios X , Humanos , Modelos Moleculares , Domínios Proteicos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa