Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 102
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Chem Inf Model ; 63(23): 7423-7443, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-37990410

RESUMO

Nuclear receptors are the fundamental building blocks of gene expression regulation and the focus of many drug targets. While binding to DNA, nuclear receptors act as transcription factors, governing a multitude of functions in the human body. Peroxisome proliferator-activator receptor γ (PPARγ) and the retinoid X receptor α (RXRα) form heterodimers with unique properties and have a primordial role in insulin sensitization. This PPARγ/RXRα heterodimer has been shown to be impacted by per- and polyfluoroalkyl substances (PFAS) and linked to a variety of significant health conditions in humans. Herein, a selection of the most common PFAS (legacy and emerging) was studied utilizing molecular dynamics simulations for PPARγ/RXRα. The local and global structural effects of PFAS binding on the known ligand binding pockets of PPARγ and RXRα as well as the DNA binding domain (DBD) of RXRα were inspected. The binding free energies were predicted computationally and were compared between the different binding pockets. In addition, two electronic structure approaches were utilized to model the interaction of PFAS within the DNA binding domain, density functional theory (DFT) and domain-based pair natural orbital coupled cluster with perturbative triples (DLPNO-CCSD(T)) approaches, with implicit solvation. Residue decomposition and hydrogen-bonding analysis were also performed, detailing the role of prominent residues in molecular recognition. The role of l-carnitine is explored as a potential in vivo remediation strategy for PFAS interaction with the PPARγ/RXRα heterodimer. In this work, it was found that PFAS can bind and act as agonists for all of the investigated pockets. For the first time in the literature, PFAS are postulated to bind to the DNA binding domain in a nonspecific manner. In addition, for the PPARγ ligand binding domain, l-carnitine shows promise in replacing smaller PFAS from the pocket.


Assuntos
Fluorocarbonos , PPAR gama , Humanos , PPAR gama/metabolismo , Ligantes , Proliferadores de Peroxissomos , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/metabolismo , DNA/química , Carnitina
2.
Eur J Med Chem ; 254: 115341, 2023 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-37058970

RESUMO

Retinoid X receptor alpha (RXRα) is an important therapeutic target of cancer. Recently, small molecules (e.g.,XS-060 and its derivatives), which can significantly induce RXRα-dependent mitotic arrest by inhibiting pRXRα-PLK1 interaction, have been demonstrated as excellent anticancer agents. To further obtain novel RXR-targeted antimitotic agents with excellent bioactivity and drug-like properties, we herein synthesized two new series of bipyridine amide derivatives with XS-060 as the lead compound. In the reporter gene assay, most synthesized compounds showed antagonistic activity against RXRα. The most active compound, bipyridine amide B9 (BPA-B9), showed better activity than XS-060, with excellent RXRα-binding affinity (KD = 39.29 ± 1.12 nM) and anti-proliferative activity against MDA-MB-231 (IC50 = 16 nM, SI > 3). Besides, a docking study revealed a proper fitting of BPA-B9 into the coactivator binding site of RXRα, rationalizing its potent antagonistic effect on RXRα transactivation. Further, the mechanism studies revealed that the anticancer activity of BPA-B9 was dependent on its cellular RXRα-targeted mechanism, such as inhibiting pRXRα-PLK1 interaction and inducing RXRα-dependent mitotic arrest. Besides, BPA-B9 displayed better pharmacokinetics than the lead XS-060. Further, animal assays indicated BPA-B9 had significant anticancer efficacy in vivo with no considerable side effects. Together, our study reveals a novel RXRα ligand BPA-B9 targeting the pRXRα-PLK1 interaction, with great potential as a promising anticancer drug candidate for further development.


Assuntos
Amidas , Antineoplásicos , Animais , Antineoplásicos/farmacologia , Sítios de Ligação , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/metabolismo
3.
J Biol Chem ; 297(1): 100814, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34081964

RESUMO

Nuclear receptors (NRs) activate transcription of target genes in response to binding of ligands to their ligand-binding domains (LBDs). Typically, in vitro assays use either gene expression or the recruitment of coactivators to the isolated LBD of the NR of interest to measure NR activation. However, this approach ignores that NRs function as homo- as well as heterodimers and that the LBD harbors the main dimerization interface. Cofactor recruitment is thereby interconnected with oligomerization status as well as ligand occupation of the partnering LBD through allosteric cross talk. Here we present a modular set of homogeneous time-resolved FRET-based assays through which we investigated the activation of PPARγ in response to ligands and the formation of heterodimers with its obligatory partner RXRα. We introduced mutations into the RXRα LBD that prevent coactivator binding but do not interfere with LBD dimerization or ligand binding. This enabled us to specifically detect PPARγ coactivator recruitment to PPARγ:RXRα heterodimers. We found that the RXRα agonist SR11237 destabilized the RXRα homodimer but promoted formation of the PPARγ:RXRα heterodimer, while being inactive on PPARγ itself. Of interest, incorporation of PPARγ into the heterodimer resulted in a substantial gain in affinity for coactivator CBP-1, even in the absence of ligands. Consequently, SR11237 indirectly promoted coactivator binding to PPARγ by shifting the oligomerization preference of RXRα toward PPARγ:RXRα heterodimer formation. These results emphasize that investigation of ligand-dependent NR activation should take NR dimerization into account. We envision these assays as the necessary assay tool kit for investigating NRs that partner with RXRα.


Assuntos
Proteína de Ligação a CREB/metabolismo , PPAR gama/metabolismo , Multimerização Proteica , Receptor X Retinoide alfa/metabolismo , Benzoatos/farmacologia , Células HEK293 , Humanos , Ligantes , Mutação/genética , Coativador 1 de Receptor Nuclear/metabolismo , PPAR gama/agonistas , PPAR gama/química , Domínios Proteicos , Multimerização Proteica/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Reprodutibilidade dos Testes , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/genética , Retinoides/farmacologia , Rosiglitazona/farmacologia , Ativação Transcricional/genética
4.
Biochemistry ; 60(15): 1165-1177, 2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33792309

RESUMO

Differential scanning calorimetry and differential scanning fluorimetry were used to measure the thermal stability of human retinoid X receptor-α ligand binding domain (RXRα LBD) homodimer in the absence or presence of rexinoid and coactivator peptide, GRIP-1. The apo-RXRα LBD homodimer displayed a single thermal unfolding transition with a Tm of 58.7 °C and an unfolding enthalpy (ΔH) of 673 kJ/mol (12.5 J/g), much lower than average value (35 J/g) of small globular proteins. Using a heat capacity change (ΔCp) of 15 kJ/(mol K) determined by measurements at different pH values, the free energy of unfolding (ΔG) of the native state was 33 kJ/mol at 37 °C. Rexinoid binding to the apo-homodimer increased Tm by 5 to 9 °C and increased the ΔG of the native homodimer by 12 to 20 kJ/mol at 37 °C, consistent with the nanomolar dissociation constant (Kd) of the rexinoids. GRIP-1 binding to holo-homodimers containing rexinoid resulted in additional increases in ΔG of 14 kJ/mol, a value that was the same for all three rexinoids. Binding of rexinoid and GRIP-1 resulted in a combined 50% increase in unfolding enthalpy, consistent with reduced structural fluidity and more compact folding observed in other published structural studies. The complexes of UAB110 and UAB111 are each more stable than the UAB30 complex by 8 kJ/mol due to enhanced hydrophobic interactions in the binding pocket because of their larger end groups. This increase in thermodynamic stability positively correlates with their improved RXR activation potency. Thermodynamic measurements are thus valuable in predicting agonist potency.


Assuntos
Peptídeos/farmacologia , Multimerização Proteica/efeitos dos fármacos , Receptor X Retinoide alfa/química , Concentração de Íons de Hidrogênio , Cinética , Estabilidade Proteica/efeitos dos fármacos , Estrutura Quaternária de Proteína/efeitos dos fármacos , Termodinâmica
5.
Dev Cell ; 55(6): 707-722.e9, 2020 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-33321102

RESUMO

Retinoid X receptor alpha (RXRα), a nuclear receptor of transcription factor, controls various physiological and pathological pathways including cellular growth, proliferation, differentiation, and apoptosis. Here, we report that RXRα is phosphorylated at its N-terminal A/B domain by cyclin-dependent kinase 1 (Cdk1) at the onset of mitosis, triggering its translocation to the centrosome, where phosphorylated-RXRα (p-RXRα) interacts with polo-like kinase 1 (PLK1) through its N-terminal A/B domain by a unique mechanism. The interaction promotes PLK1 activation, centrosome maturation, and mitotic progression. Levels of p-RXRα are abnormally elevated in cancer cell lines, during carcinogenesis in animals, and in clinical tumor tissues. An RXRα ligand XS060, which specifically inhibits p-RXRα/PLK1 interaction but not RXRα heterodimerization, promotes mitotic arrest and catastrophe in a tumor-specific manner. These findings unravel a transcription-independent action of RXRα at the centrosome during mitosis and identify p-RXRα as a tumor-specific vulnerability for developing mitotic drugs with improved therapeutic index.


Assuntos
Carcinogênese/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrossomo/metabolismo , Mitose , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptor X Retinoide alfa/metabolismo , Animais , Sítios de Ligação , Proteína Quinase CDC2/metabolismo , Carcinogênese/patologia , Proteínas de Ciclo Celular/química , Feminino , Células HeLa , Células Hep G2 , Humanos , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ligação Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Proto-Oncogênicas/química , Receptor X Retinoide alfa/química
6.
Nucleic Acids Res ; 48(19): 11199-11213, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-32990725

RESUMO

The MED1 subunit of the Mediator complex is an essential coactivator of nuclear receptor-mediated transcriptional activation. While structural requirements for ligand-dependent binding of classical coactivator motifs of MED1 to numerous nuclear receptor ligand-binding domains have been fully elucidated, the recognition of the full-length or truncated coactivator by full nuclear receptor complexes remain unknown. Here we present structural details of the interaction between a large part of MED1 comprising its structured N-terminal and the flexible receptor-interacting domains and the mutual heterodimer of the vitamin D receptor (VDR) and the retinoid X receptor (RXR) bound to their cognate DNA response element. Using a combination of structural and biophysical methods we show that the ligand-dependent interaction between VDR and the second coactivator motif of MED1 is crucial for complex formation and we identify additional, previously unseen, interaction details. In particular, we identified RXR regions involved in the interaction with the structured N-terminal domain of MED1, as well as VDR regions outside the classical coactivator binding cleft affected by coactivator recruitment. These findings highlight important roles of each receptor within the heterodimer in selective recognition of MED1 and contribute to our understanding of the nuclear receptor-coregulator complexes.


Assuntos
DNA/metabolismo , Subunidade 1 do Complexo Mediador , Receptores de Calcitriol , Receptor X Retinoide alfa , Humanos , Ligantes , Subunidade 1 do Complexo Mediador/química , Subunidade 1 do Complexo Mediador/metabolismo , Ligação Proteica , Domínios Proteicos , Multimerização Proteica , Receptores de Calcitriol/química , Receptores de Calcitriol/metabolismo , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/metabolismo
7.
Theranostics ; 9(19): 5444-5463, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31534496

RESUMO

PPARγ is a pharmacological target in inflammatory and metabolic diseases. Upon agonistic treatment or following antagonism, binding of co-factors is altered, which consequently affects PPARγ-dependent transactivation as well as its DNA-independent properties. Therefore, establishing techniques to characterize these interactions is an important issue in living cells. Methods: Using the FRET pair Clover/mRuby2, we set up a flow cytometry-based FRET assay by analyzing PPARγ1 binding to its heterodimerization partner RXRα. Analyses of PPARγ-reporter and co-localization studies by laser-scanning microscopy validated this system. Refining the system, we created a new readout to distinguish strong from weak interactions, focusing on PPARγ-binding to the co-repressor N-CoR2. Results: We observed high FRET in cells expressing Clover-PPARγ1 and mRuby2-RXRα, but no FRET when cells express a mRuby2-RXRα deletion mutant, lacking the PPARγ interaction domain. Focusing on the co-repressor N-CoR2, we identified in HEK293T cells the new splice variant N-CoR2-ΔID1-exon. Overexpressing this isoform tagged with mRuby2, revealed no binding to Clover-PPARγ1, nor in murine J774A.1 macrophages. In HEK293T cells, binding was even lower in comparison to N-CoR2 constructs in which domains established to mediate interaction with PPARγ binding are deleted. These data suggest a possible role of N-CoR2-ΔID1-exon as a dominant negative variant. Because binding to N-CoR2-mRuby2 was not altered following activation or antagonism of Clover-PPARγ1, we determined the effect of pharmacological treatment on FRET intensity. Therefore, we calculated flow cytometry-based FRET efficiencies based on our flow cytometry data. As with PPARγ antagonism, PPARγ agonist treatment did not prevent binding of N-CoR2. Conclusion: Our system allows the close determination of protein-protein interactions with a special focus on binding intensity, allowing this system to characterize the role of protein domains as well as the effect of pharmacological agents on protein-protein interactions.


Assuntos
Citometria de Fluxo/métodos , Transferência Ressonante de Energia de Fluorescência/métodos , PPAR gama/metabolismo , Animais , Dimerização , Células HEK293 , Humanos , Camundongos , Correpressor 1 de Receptor Nuclear/química , Correpressor 1 de Receptor Nuclear/genética , Correpressor 1 de Receptor Nuclear/metabolismo , PPAR gama/química , PPAR gama/genética , Ligação Proteica , Domínios Proteicos , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/metabolismo
8.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 2): 98-104, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30713160

RESUMO

The retinoic X receptor (RXR) plays a crucial role in the superfamily of nuclear receptors (NRs) by acting as an obligatory partner of several nuclear receptors; its role as a transcription factor is thus critical in many signalling pathways, such as metabolism, cell development, differentiation and cellular death. The first published structure of the apo ligand-binding domain (LBD) of RXRα, which is still used as a reference today, contained inaccuracies. In the present work, these inaccuracies were corrected using modern crystallographic tools. The most important correction concerns the presence of a π-bulge in helix H7, which was originally built as a regular α-helix. The presence of several CHAPS molecules, which are visible for the first time in the electron-density map and which stabilize the H1-H3 loop, which contains helix H2, are also revealed. The apo RXR structure has played an essential role in deciphering the molecular mode of action of NR ligands and is still used in numerous biophysical studies. This refined structure should be used preferentially in the future in interpreting experiments as well as for modelling and structural dynamics studies of the apo RXRα LBD.


Assuntos
Apoproteínas/química , Apoproteínas/metabolismo , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalização , Cristalografia por Raios X , Humanos , Ligantes , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Domínios Proteicos
9.
J Sep Sci ; 42(3): 744-753, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30488556

RESUMO

Employing immobilized metal-ion affinity chromatography and magnetic separation could ideally provide a useful analytical strategy for purifying His-tagged protein. In the current study, a facile route was designed to prepare CMPEI-Ni2+ @SiO2 @Fe3 O4 (CMPEI=carboxymethylated polyethyleneimine) magnetic nanoparticles composed of a strong magnetic core of Fe3 O4 and a Ni2+ -immobilized carboxymethylated polyethyleneimine coated outside shell, which was formed by electrostatic interactions between polyanionic electrolyte of carboxymethylated polyethyleneimine and positively charged surface of 3-(trimethoxysilyl)propylamin modified SiO2 @Fe3 O4 . The resulting CMPEI-Ni2+ @SiO2 @Fe3 O4 composite nanoparticles displayed well-uniform structure and high magnetic responsiveness. Hexa His-tagged peptides and purified His-tagged recombinant retinoid X receptor alpha were chosen as the model samples to evaluate the adsorption, capacity, and reusability of the composite nanoparticles. The results demonstrated the CMPEI-Ni2+ @SiO2 @Fe3 O4 nanoparticles possessed rapid adsorption, large capacity, and good recyclability. The obtained nanoparticles were further used to purify His-tagged protein in practical environment. It was found that the nanoparticles could selectively capture His-tagged recombinant retinoid X receptor protein from complex cell lysate. Owing to its easy synthesis, large binding capacity, and good reusability, the prepared CMPEI-Ni2+ @SiO2 @Fe3 O4 magnetic nanoparticles have great potential for application in biotechnological fields.


Assuntos
Histidina/química , Nanopartículas de Magnetita/química , Polietilenoimina/química , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/isolamento & purificação , Adsorção , Histidina/isolamento & purificação , Estrutura Molecular , Tamanho da Partícula , Propriedades de Superfície
10.
FEBS Lett ; 593(2): 242-250, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30565665

RESUMO

1-[(3,5,5,8,8-pentamethyl-5,6,7,8-tetrahydronaphthalen-2-yl)amino]benzotriazole-5-carboxylic acid (CBt-PMN), a partial agonist of retinoid X receptor (RXR), has attracted attention due to its potential to treat type 2 diabetes and central nervous system diseases with reduced adverse effects of existing full agonists. Herein, we report the crystal structure of CBt-PMN-bound ligand-binding domain of human RXRα (hRXRα) and its biochemical characterization. Interestingly, the structure is a tetramer in nature, in which CBt-PMNs are clearly found binding in two different conformations. The dynamics of the hRXRα/CBt-PMN complex examined using molecular dynamics simulations suggest that the flexibility of the AF-2 interface depends on the conformation of the ligand. These facts reveal that the dual conformation of CBt-PMN in the complex is probably the reason behind its partial agonistic activity.


Assuntos
Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/metabolismo , Tetra-Hidronaftalenos/química , Tetra-Hidronaftalenos/farmacologia , Triazóis/química , Triazóis/farmacologia , Sítios de Ligação , Cristalografia por Raios X , Humanos , Ligantes , Modelos Moleculares , Conformação Molecular , Simulação de Dinâmica Molecular , Ligação Proteica
11.
J Biol Chem ; 293(47): 18180-18191, 2018 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-30275017

RESUMO

Nuclear receptor farnesoid X receptor (FXR) functions as the major bile acid sensor coordinating cholesterol metabolism, lipid homeostasis, and absorption of dietary fats and vitamins. Because of its central role in metabolism, FXR represents an important drug target to manage metabolic and other diseases, such as primary biliary cirrhosis and nonalcoholic steatohepatitis. FXR and nuclear receptor retinoid X receptor α (RXRα) form a heterodimer that controls the expression of numerous downstream genes. To date, the structural basis and functional consequences of the FXR/RXR heterodimer interaction have remained unclear. Herein, we present the crystal structures of the heterodimeric complex formed between the ligand-binding domains of human FXR and RXRα. We show that both FXR and RXR bind to the transcriptional coregulator steroid receptor coactivator 1 with higher affinity when they are part of the heterodimer complex than when they are in their respective monomeric states. Furthermore, structural comparisons of the FXR/RXRα heterodimers and the FXR monomers bound with different ligands indicated that both heterodimerization and ligand binding induce conformational changes in the C terminus of helix 11 in FXR that affect the stability of the coactivator binding surface and the coactivator binding in FXR. In summary, our findings shed light on the allosteric signal transduction in the FXR/RXR heterodimer, which may be utilized for future drug development targeting FXR.


Assuntos
Coativador 1 de Receptor Nuclear/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/metabolismo , Cristalografia por Raios X , Dimerização , Humanos , Ligantes , Coativador 1 de Receptor Nuclear/química , Coativador 1 de Receptor Nuclear/genética , Ligação Proteica , Domínios Proteicos , Receptores Citoplasmáticos e Nucleares/genética , Receptor X Retinoide alfa/genética
12.
Cell Mol Biol Lett ; 23: 36, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30093910

RESUMO

The nuclear receptor RXRα (retinoid X receptor-α) is a transcription factor that regulates the expression of multiple genes. Its non-genomic function is largely related to its structure, polymeric forms and modification. Previous research revealed that some non-genomic activity of RXRα occurs via formation of heterodimers with Nur77. RXRα-Nur77 heterodimers translocate from the nucleus to the mitochondria in response to certain apoptotic stimuli and this activity correlates with cell apoptosis. More recent studies revealed a significant role for truncated RXRα (tRXRα), which interacts with the p85α subunit of the PI3K/AKT signaling pathway, leading to enhanced activation of AKT and promoting cell growth in vitro and in animals. We recently reported on a series of NSAID sulindac analogs that can bind to tRXRα through a unique binding mechanism. We also identified one analog, K-80003, which can inhibit cancer cell growth by inducing tRXRα to form a tetramer, thus disrupting p85α-tRXRα interaction. This review analyzes the non-genomic effects of RXRα in normal and tumor cells, and discusses the functional differences based on RXRα protein structure (structure source: the RCSB Protein Data Bank).


Assuntos
Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/metabolismo , Animais , Sítios de Ligação , Bases de Dados de Proteínas , Descoberta de Drogas , Humanos , Modelos Moleculares , Neoplasias/metabolismo , Conformação Proteica , Multimerização Proteica
13.
Nat Commun ; 8(1): 868, 2017 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-29021580

RESUMO

Assessing the physical connections and allosteric communications in multi-domain nuclear receptor (NR) polypeptides has remained challenging, with few crystal structures available to show their overall structural organizations. Here we report the quaternary architecture of multi-domain retinoic acid receptor ß-retinoic X receptor α (RARß-RXRα) heterodimer bound to DNA, ligands and coactivator peptides, examined through crystallographic, hydrogen-deuterium exchange mass spectrometry, mutagenesis and functional studies. The RARß ligand-binding domain (LBD) and DNA-binding domain (DBD) are physically connected to foster allosteric signal transmission between them. Direct comparisons among all the multi-domain NRs studied crystallographically to date show significant variations within their quaternary architectures, rather than a common architecture adhering to strict rules. RXR remains flexible and adaptive by maintaining loosely organized domains, while its heterodimerization partners use a surface patch on their LBDs to form domain-domain interactions with DBDs.Nuclear receptors (NR) are multidomain proteins, which makes their crystallization challenging. Here the authors present the crystal structure of the retinoic acid receptor ß-retinoic X receptor α (RARß-RXRα) heterodimer bound to DNA, ligands and coactivator peptides, which shows that NR quaternary architectures are variable.


Assuntos
Receptores do Ácido Retinoico/química , Receptor X Retinoide alfa/química , Regulação Alostérica , Células HEK293 , Humanos , Estrutura Quaternária de Proteína
14.
Structure ; 25(10): 1506-1518.e4, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28890360

RESUMO

Nuclear receptor (NR) transcription factors bind various coreceptors, small-molecule ligands, DNA response element sequences, and transcriptional coregulator proteins to affect gene transcription. Small-molecule ligands and DNA are known to influence receptor structure, coregulator protein interaction, and function; however, little is known on the mechanism of synergy between ligand and DNA. Using quantitative biochemical, biophysical, and solution structural methods, including 13C-detected nuclear magnetic resonance and hydrogen/deuterium exchange (HDX) mass spectrometry, we show that ligand and DNA cooperatively recruit the intrinsically disordered steroid receptor coactivator-2 (SRC-2/TIF2/GRIP1/NCoA-2) receptor interaction domain to peroxisome proliferator-activated receptor gamma-retinoid X receptor alpha (PPARγ-RXRα) heterodimer and reveal the binding determinants of the complex. Our data reveal a thermodynamic mechanism by which DNA binding propagates a conformational change in PPARγ-RXRα, stabilizes the receptor ligand binding domain dimer interface, and impacts ligand potency and cooperativity in NR coactivator recruitment.


Assuntos
DNA/metabolismo , Complexos Multiproteicos/química , Coativador 2 de Receptor Nuclear/química , Coativador 2 de Receptor Nuclear/metabolismo , Sítios de Ligação , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13 , Medição da Troca de Deutério , Regulação da Expressão Gênica , Humanos , Ligantes , PPAR gama/química , PPAR gama/metabolismo , Ligação Proteica , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/metabolismo
15.
J Mol Graph Model ; 77: 121-129, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28850894

RESUMO

The treatment of many diseases may require drugs that are capable to attack multiple targets simultaneously. Obviously, the virtual screening of multi-target drug candidates is much more time consuming compared to the single-target case. This, in particular, concerns the last step of virtual screening where the binding free energy is computed by conventional molecular dynamics simulation. To overcome this difficulty we propose a simple protocol which is relied on the fast steered molecular dynamics simulation and on available experimental data on binding affinity of reference ligand to a given target. Namely, first we compute non-equilibrium works generated during pulling ligands from the binding site using the steered molecular dynamics method. Then as top leads we choose only those compounds that have the non-equilibrium work larger than that of a reference compound for which the binding free energy has been already known from experiment. Despite many efforts no cures for AD (Alzheimer's disease) have been found. One of possible reasons for this failure is that drug candidates were developed for a single target, while there are exist many possible pathways to AD. Applying our new protocol to five targets including amyloid beta fibril, peroxisome proliferator-activated receptor γ, retinoic X receptor α, ß- and γ-secretases, we have found two potential drugs (CID 16040294 and CID 9998128) for AD from the large PubChem database. We have also shown that these two ligands can interfere with the activity of popular Acetylcholinesterase target through strong binding towards it.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Interface Usuário-Computador , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/química , Peptídeos beta-Amiloides/antagonistas & inibidores , Peptídeos beta-Amiloides/química , Sítios de Ligação , Sistemas de Liberação de Medicamentos , Humanos , Ligantes , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Receptor X Retinoide alfa/antagonistas & inibidores , Receptor X Retinoide alfa/química
16.
Nat Commun ; 8(1): 103, 2017 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-28740126

RESUMO

Muscle-invasive bladder cancer (MIBC) is an aggressive disease with limited therapeutic options. Although immunotherapies are approved for MIBC, the majority of patients fail to respond, suggesting existence of complementary immune evasion mechanisms. Here, we report that the PPARγ/RXRα pathway constitutes a tumor-intrinsic mechanism underlying immune evasion in MIBC. Recurrent mutations in RXRα at serine 427 (S427F/Y), through conformational activation of the PPARγ/RXRα heterodimer, and focal amplification/overexpression of PPARγ converge to modulate PPARγ/RXRα-dependent transcription programs. Immune cell-infiltration is controlled by activated PPARγ/RXRα that inhibits expression/secretion of inflammatory cytokines. Clinical data sets and an in vivo tumor model indicate that PPARγHigh/RXRαS427F/Y impairs CD8+ T-cell infiltration and confers partial resistance to immunotherapies. Knockdown of PPARγ or RXRα and pharmacological inhibition of PPARγ significantly increase cytokine expression suggesting therapeutic approaches to reviving immunosurveillance and sensitivity to immunotherapies. Our study reveals a class of tumor cell-intrinsic "immuno-oncogenes" that modulate the immune microenvironment of cancer.Muscle-invasive bladder cancer (MIBC) is a potentially lethal disease. Here the authors characterize diverse genetic alterations in MIBC that convergently lead to constitutive activation of PPARgamma/RXRalpha and result in immunosurveillance escape by inhibiting CD8+ T-cell recruitment.


Assuntos
Evasão da Resposta Imune/imunologia , Monitorização Imunológica , PPAR gama/imunologia , Receptor X Retinoide alfa/imunologia , Neoplasias da Bexiga Urinária/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/imunologia , Citocinas/metabolismo , Perfilação da Expressão Gênica/métodos , Células HCT116 , Humanos , Immunoblotting , Imunoterapia/métodos , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Camundongos , Microscopia de Fluorescência , Mutação/imunologia , Invasividade Neoplásica , PPAR gama/química , PPAR gama/genética , Multimerização Proteica/imunologia , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/genética , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/terapia
17.
ACS Chem Neurosci ; 8(9): 2065-2077, 2017 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-28691794

RESUMO

Retinoid X receptors (RXRs) play key roles in many physiological processes in both the periphery and central nervous system. In addition, RXRs form heterodimers with other nuclear receptors to exert their physiological effects. The nuclear receptor related 1 protein (NURR1) is particularly interesting because of its role in promoting differentiation and survival of dopamine neurons. However, only a small number of RXR-heterodimer selective modulators are available, with limited chemical diversity. This work describes the synthesis, biochemical evaluation, and structural elucidation of a novel series of RXR ligands with strongly biased interactions with RXRα-NURR1 heterodimers. Targeted modifications to the small molecule biaryl scaffold caused local RXRα side-chain disturbances and displacement of secondary structural elements upon ligand binding. This resulted in the repositioning of protein helices in the heterodimer interface of RXRα, alterations in homo- versus heterodimer formation, and modulation of activation function 2 (AF2). The data provide a rationale for the design of RXR ligands consisting of a highly conserved hydrophilic region, strongly contributing to the ligand affinity, and a variable hydrophobic region, which efficiently probes the effects of structural changes at the level of the ligand on co-regulator recruitment or the RXRα-NURR1 dimerization interface.


Assuntos
Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/química , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/metabolismo , Desenho de Fármacos , Escherichia coli , Ésteres/química , Éteres/química , Humanos , Ligação de Hidrogênio , Ligantes , Modelos Moleculares , Estrutura Molecular , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/agonistas , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/antagonistas & inibidores , Ligação Proteica , Multimerização Proteica , Receptor X Retinoide alfa/agonistas , Receptor X Retinoide alfa/antagonistas & inibidores , Técnicas do Sistema de Duplo-Híbrido
18.
Angew Chem Int Ed Engl ; 56(20): 5480-5484, 2017 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-28407400

RESUMO

Spiroketals are structural motifs found in many biologically active natural products, which has stimulated considerable efforts toward their synthesis and interest in their use as drug lead compounds. Despite this, the use of spiroketals, and especially bisbenzanulated spiroketals, in a structure-based drug discovery setting has not been convincingly demonstrated. Herein, we report the rational design of a bisbenzannulated spiroketal that potently binds to the retinoid X receptor (RXR) thereby inducing partial co-activator recruitment. We solved the crystal structure of the spiroketal-hRXRα-TIF2 ternary complex, and identified a canonical allosteric mechanism as a possible explanation for the partial agonist behavior of our spiroketal. Our co-crystal structure, the first of a designed spiroketal-protein complex, suggests that spiroketals can be designed to selectively target other nuclear receptor subtypes.


Assuntos
Furanos/química , Coativador 2 de Receptor Nuclear/química , Receptor X Retinoide alfa/química , Compostos de Espiro/química , Cristalografia por Raios X , Humanos , Modelos Moleculares , Estrutura Molecular
19.
Proc Natl Acad Sci U S A ; 114(15): 3999-4004, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28348207

RESUMO

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the loss of dopaminergic (DAergic) neurons in the substantia nigra and the gradual depletion of dopamine (DA). Current treatments replenish the DA deficit and improve symptoms but induce dyskinesias over time, and neuroprotective therapies are nonexistent. Here we report that Nuclear receptor-related 1 (Nurr1):Retinoid X receptor α (RXRα) activation has a double therapeutic potential for PD, offering both neuroprotective and symptomatic improvement. We designed BRF110, a unique in vivo active Nurr1:RXRα-selective lead molecule, which prevents DAergic neuron demise and striatal DAergic denervation in vivo against PD-causing toxins in a Nurr1-dependent manner. BRF110 also protects against PD-related genetic mutations in patient induced pluripotent stem cell (iPSC)-derived DAergic neurons and a genetic mouse PD model. Remarkably, besides neuroprotection, BRF110 up-regulates tyrosine hydroxylase (TH), aromatic l-amino acid decarboxylase (AADC), and GTP cyclohydrolase I (GCH1) transcription; increases striatal DA in vivo; and has symptomatic efficacy in two postneurodegeneration PD models, without inducing dyskinesias on chronic daily treatment. The combined neuroprotective and symptomatic effects of BRF110 identify Nurr1:RXRα activation as a potential monotherapeutic approach for PD.


Assuntos
Antiparkinsonianos/farmacologia , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Doença de Parkinson/tratamento farmacológico , Receptor X Retinoide alfa/metabolismo , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Antiparkinsonianos/química , Antiparkinsonianos/farmacocinética , Encéfalo/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Dopamina/genética , Estabilidade de Medicamentos , Humanos , Masculino , Camundongos Endogâmicos BALB C , Terapia de Alvo Molecular , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/fisiologia , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/agonistas , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Multimerização Proteica , Ratos , Receptor X Retinoide alfa/agonistas , Receptor X Retinoide alfa/química , Receptor X Retinoide alfa/genética
20.
J Mol Model ; 23(3): 98, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28251414

RESUMO

Bexarotene is an FDA approved retinoid X-receptor (RXR) agonist for the treatment of cutaneous T-cell lymphoma, and its use in other cancers and Alzheimer's disease is being investigated. The drug causes serious side effects, which might be reduced by chemical modifications of the molecule. To rationalize known agonists and to help identify sites for potential substitutions we present molecular simulations in which the RXR ligand-binding domain was flooded with a large number of drug-like molecules, and molecular dynamics simulations of a series of bexarotene-like ligands bound to the RXR ligand-binding domain. Based on the flooding simulations, two regions of interest for ligand modifications were identified: a hydrophobic area near the bridgehead and another near the fused ring. In addition, positional fluctuations of the phenyl ring were generally smaller than fluctuations of the fused ring of the ligands. Together, these observations suggest that the fused ring might be a good target for the design of higher affinity bexarotene-like ligands, while the phenyl ring is already optimized. In addition, notable differences in ligand position and interactions between the RXRα and RXRß were observed, as well as differences in hydrogen bonding and solvation, which might be exploited in the development of subspecies-specific ligands.


Assuntos
Receptor X Retinoide alfa/química , Receptor X Retinoide beta/química , Tetra-Hidronaftalenos/química , Bexaroteno , Sítios de Ligação , Humanos , Ligação de Hidrogênio , Ligantes , Simulação de Dinâmica Molecular , Ligação Proteica , Receptor X Retinoide alfa/agonistas , Receptor X Retinoide beta/agonistas , Tetra-Hidronaftalenos/efeitos adversos , Tetra-Hidronaftalenos/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...