Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Chem Biol ; 26(5): 674-685.e6, 2019 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-30853418

RESUMO

Nurr1, a nuclear receptor essential for the development, maintenance, and survival of midbrain dopaminergic neurons, is a potential therapeutic target for Parkinson's disease, a neurological disorder characterized by the degeneration of these same neurons. Efforts to identify Nurr1 agonists have been hampered by the recognition that it lacks several classic regulatory elements of nuclear receptor function, including the canonical ligand-binding pocket. Here we report that the dopamine metabolite 5,6-dihydroxyindole (DHI) binds directly to and modulates the activity of Nurr1. Using biophysical assays and X-ray crystallography, we show that DHI binds to the ligand-binding domain within a non-canonical pocket, forming a covalent adduct with Cys566. In cultured cells and zebrafish, DHI stimulates Nurr1 activity, including the transcription of target genes underlying dopamine homeostasis. These findings suggest avenues for developing synthetic Nurr1 ligands to ameliorate the symptoms and progression of Parkinson's disease.


Assuntos
Dopamina/metabolismo , Indóis/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , Dopamina/química , Humanos , Indóis/química , Indóis/farmacologia , Larva/metabolismo , Simulação de Dinâmica Molecular , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/química , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Estresse Oxidativo/efeitos dos fármacos , Domínios Proteicos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Termodinâmica , Transcrição Gênica/efeitos dos fármacos , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
2.
Nat Commun ; 9(1): 4055, 2018 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-30305617

RESUMO

Epithelial dysfunction and crypt destruction are defining features of inflammatory bowel disease (IBD). However, current IBD therapies targeting epithelial dysfunction are lacking. The nuclear receptor LRH-1 (NR5A2) is expressed in intestinal epithelium and thought to contribute to epithelial renewal. Here we show that LRH-1 maintains intestinal epithelial health and protects against inflammatory damage. Knocking out LRH-1 in murine intestinal organoids reduces Notch signaling, increases crypt cell death, distorts the cellular composition of the epithelium, and weakens the epithelial barrier. Human LRH-1 (hLRH-1) rescues epithelial integrity and when overexpressed, mitigates inflammatory damage in murine and human intestinal organoids, including those derived from IBD patients. Finally, hLRH-1 greatly reduces disease severity in T-cell-mediated murine colitis. Together with the failure of a ligand-incompetent hLRH-1 mutant to protect against TNFα-damage, these findings provide compelling evidence that hLRH-1 mediates epithelial homeostasis and is an attractive target for intestinal disease.


Assuntos
Epitélio/patologia , Homeostase , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Diferenciação Celular , Sobrevivência Celular , Colite/metabolismo , Colite/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Organoides/metabolismo , Receptores Notch/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
3.
ACS Chem Biol ; 12(12): 2934-2939, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-28981251

RESUMO

Resistance to clinical antiandrogens has plagued the evolution of effective therapeutics for advanced prostate cancer. As with the first-line therapeutic bicalutamide (Casodex), resistance to newer antiandrogens (enzalutamide, ARN-509) develops quickly in patients, despite the fact that these drugs have ∼10-fold better affinity for the androgen receptor than bicalutamide. Improving affinity alone is often not sufficient to prevent resistance, and alternative strategies are needed to improve antiandrogen efficacy. Covalent and reversible covalent drugs are being used to thwart drug resistance in other contexts, and activated aryl nitriles are among the moieties being exploited for this purpose. We capitalized on the presence of an aryl nitrile in bicalutamide, and the existence of a native cysteine residue (Cys784) in the androgen receptor ligand binding pocket, to develop 5N-bicalutamide, a cysteine-reactive antiandrogen. 5N-bicalutamide exhibits a 150-fold improvement in Ki and 20-fold improvement in IC50 over the parent compound. We attribute the marked improvement in affinity and activity to the formation of a covalent adduct with Cys784, a residue that is not among the more than 160 androgen receptor point mutations associated with prostate cancer. Increasing the residence time of bound antiandrogen via formation of a covalent adduct may forestall the drug resistance seen with current clinical antiandrogens.


Assuntos
Antagonistas de Androgênios/química , Antagonistas de Androgênios/farmacologia , Anilidas/farmacologia , Nitrilas/farmacologia , Compostos de Tosil/farmacologia , Anilidas/química , Sítios de Ligação , Células HeLa , Humanos , Modelos Moleculares , Estrutura Molecular , Nitrilas/química , Ligação Proteica , Conformação Proteica , Receptores Androgênicos/química , Receptores Androgênicos/metabolismo , Relação Estrutura-Atividade , Compostos de Tosil/química
4.
PLoS One ; 11(7): e0159316, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27467220

RESUMO

Conventional efforts relying on high-throughput physical and virtual screening of large compound libraries have failed to yield high-efficiency chemical probes for many of the 48 human nuclear receptors. Here, we investigated whether disulfide-trapping, an approach new to nuclear receptors, would provide effective lead compounds targeting human liver receptor homolog 1 (hLRH-1, NR5A2). Despite the fact that hLRH-1 contains a large ligand binding pocket and binds phospholipids with high affinity, existing synthetic hLRH-1 ligands are of limited utility due to poor solubility, low efficacy or significant off-target effects. Using disulfide-trapping, we identified a lead compound that conjugates with remarkably high-efficiency to a native cysteine residue (Cys346) lining the hydrophobic cavity in the ligand binding domain of hLRH-1. Guided by computational modeling and cellular assays, the lead compound was elaborated into ligands PME8 and PME9 that bind hLRH-1 reversibly (no cysteine reactivity) and increase hLRH-1 activity in cells. When compared with the existing hLRH-1 synthetic agonist RJW100, both PME8 and PME9 showed comparable induction of the LRH-1 dependent target gene CYP24A1 in human HepG2 cells, beginning as early as 3 h after drug treatment. The induction is specific as siRNA-mediated knock-down of hLRH-1 renders both PME8 and PME9 ineffective. These data show that PME8 and PME9 are potent activators of hLRH-1 and suggest that with further development this lead series may yield useful chemical probes for manipulating LRH-1 activity in vivo.


Assuntos
Dissulfetos/química , Sondas Moleculares/química , Receptores Citoplasmáticos e Nucleares/química , Células Hep G2 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Simulação de Acoplamento Molecular
5.
J Mol Biol ; 428(6): 1130-1141, 2016 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-26829219

RESUMO

The Dlx5 homeodomain is a transcription factor related to the Drosophila distal-less gene that is associated with breast and lung cancer, lymphoma, Rett syndrome and osteoporosis in humans. Mutations in the DLX5 gene have been linked to deficiencies in craniofacial and limb development in higher eukaryotes, including split hand and foot malformation 1 in humans. Our characterization of a Dlx5 homeodomain:(CGACTAATTAGTCG)2 complex by NMR spectroscopy paved the way for determination of its crystal structure at 1.85Å resolution that enabled rationalization of the effects of disease-related mutations on the protein function. A Q186H mutation linked to split hand and foot malformation 1 likely affects affinity of DNA binding by disrupting water-mediated interactions with the DNA major groove. A more subtle effect is implicated for the Q178P mutation, which is not in direct contact with the DNA. Our data indicate that these mutations diminish the ability of the Dlx5 homeodomain to recognize and bind target DNAs, and they likely destabilize the formation of functional complexes.


Assuntos
DNA/química , DNA/metabolismo , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Deformidades Congênitas dos Membros/genética , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Cristalografia por Raios X , Proteínas de Homeodomínio/genética , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Conformação de Ácido Nucleico , Ligação Proteica , Conformação Proteica , Fatores de Transcrição/genética
6.
Elife ; 42015 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-26653140

RESUMO

SUMO-modification of nuclear proteins has profound effects on gene expression. However, non-toxic chemical tools that modulate sumoylation in cells are lacking. Here, to identify small molecule sumoylation inhibitors we developed a cell-based screen that focused on the well-sumoylated substrate, human Liver Receptor Homolog-1 (hLRH-1, NR5A2). Our primary gene-expression screen assayed two SUMO-sensitive transcripts, APOC3 and MUC1, that are upregulated by SUMO-less hLRH-1 or by siUBC9 knockdown, respectively. A polyphenol, tannic acid (TA) emerged as a potent sumoylation inhibitor in vitro (IC50 = 12.8 µM) and in cells. TA also increased hLRH-1 occupancy on SUMO-sensitive transcripts. Most significantly, when tested in humanized mouse primary hepatocytes, TA inhibits hLRH-1 sumoylation and induces SUMO-sensitive genes, thereby recapitulating the effects of expressing SUMO-less hLRH-1 in mouse liver. Our findings underscore the benefits of phenotypic screening for targeting post-translational modifications, and illustrate the potential utility of TA for probing the cellular consequences of sumoylation.


Assuntos
Inibidores Enzimáticos/isolamento & purificação , Inibidores Enzimáticos/metabolismo , Hepatócitos/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Sumoilação/efeitos dos fármacos , Taninos/isolamento & purificação , Taninos/metabolismo , Animais , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/métodos , Perfilação da Expressão Gênica , Hepatócitos/enzimologia , Humanos , Concentração Inibidora 50 , Camundongos , Camundongos SCID
7.
J Struct Biol ; 192(3): 342-348, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26416531

RESUMO

The nuclear receptor LRH-1 (Liver Receptor Homolog-1, NR5A2) is a transcription factor that regulates gene expression programs critical for many aspects of metabolism and reproduction. Although LRH-1 is able to bind phospholipids, it is still considered an orphan nuclear receptor (NR) with an unknown regulatory hormone. Our prior cellular and structural studies demonstrated that the signaling phosphatidylinositols PI(4,5)P2 (PIP2) and PI(3,4,5)P3 (PIP3) bind and regulate SF-1 (Steroidogenic Factor-1, NR5A1), a close homolog of LRH-1. Here, we describe the crystal structure of human LRH-1 ligand binding domain (LBD) bound by PIP3 - the first phospholipid with a head group endogenous to mammals. We show that the phospholipid hormone binds LRH-1 with high affinity, stabilizing the receptor LBD. While the hydrophobic PIP3 tails (C16/C16) are buried inside the LRH-1 ligand binding pocket, the negatively charged PIP3 head group is presented on the receptor surface, similar to the phosphatidylinositol binding mode observed in the PIP3-SF-1 structure. Thus, data presented in this work reinforce our earlier findings demonstrating that signaling phosphatidylinositols regulate the NR5A receptors LRH-1 and SF-1.


Assuntos
Fosfatidilinositóis/química , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/ultraestrutura , Fator Esteroidogênico 1/ultraestrutura , Sítios de Ligação/fisiologia , Cristalografia por Raios X , Receptor Nuclear Órfão DAX-1/química , Humanos , Modelos Moleculares , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína , Fator Esteroidogênico 1/química
8.
PLoS One ; 10(7): e0134015, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26207810

RESUMO

FKBP52 and ß-catenin have emerged in recent years as attractive targets for prostate cancer treatment. ß-catenin interacts directly with the androgen receptor (AR) and has been characterized as a co-activator of AR-mediated transcription. FKBP52 is a positive regulator of AR in cellular and whole animal models and is required for the development of androgen-dependent tissues. We previously characterized an AR inhibitor termed MJC13 that putatively targets the AR BF3 surface to specifically inhibit FKBP52-regulated AR signaling. Predictive modeling suggests that ß-catenin interacts with the AR hormone binding domain on a surface that overlaps with BF3. Here we demonstrate that FKBP52 and ß-catenin interact directly in vitro and act in concert to promote a synergistic up-regulation of both hormone-independent and -dependent AR signaling. Our data demonstrate that FKBP52 promotes ß-catenin interaction with AR and is required for ß-catenin co-activation of AR activity in prostate cancer cells. MJC13 effectively blocks ß-catenin interaction with the AR LBD and the synergistic up-regulation of AR by FKBP52 and ß-catenin. Our data suggest that co-regulation of AR by FKBP52 and ß-catenin does not require FKBP52 PPIase catalytic activity, nor FKBP52 binding to Hsp90. However, the FKBP52 proline-rich loop that overhangs the PPIase pocket is critical for synergy.


Assuntos
Receptores Androgênicos/metabolismo , Sistemas do Segundo Mensageiro , Proteínas de Ligação a Tacrolimo/metabolismo , beta Catenina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Células HEK293 , Células HeLa , Humanos , Dados de Sequência Molecular , Ligação Proteica , Proteínas de Ligação a Tacrolimo/química , beta Catenina/química
9.
Proc Natl Acad Sci U S A ; 112(15): 4666-71, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25825768

RESUMO

NANOG (from Irish mythology Tír na nÓg) transcription factor plays a central role in maintaining pluripotency, cooperating with OCT4 (also known as POU5F1 or OCT3/4), SOX2, and other pluripotency factors. Although the physiological roles of the NANOG protein have been extensively explored, biochemical and biophysical properties in relation to its structural analysis are poorly understood. Here we determined the crystal structure of the human NANOG homeodomain (hNANOG HD) bound to an OCT4 promoter DNA, which revealed amino acid residues involved in DNA recognition that are likely to be functionally important. We generated a series of hNANOG HD alanine substitution mutants based on the protein-DNA interaction and evolutionary conservation and determined their biological activities. Some mutant proteins were less stable, resulting in loss or decreased affinity for DNA binding. Overexpression of the orthologous mouse NANOG (mNANOG) mutants failed to maintain self-renewal of mouse embryonic stem cells without leukemia inhibitory factor. These results suggest that these residues are critical for NANOG transcriptional activity. Interestingly, one mutant, hNANOG L122A, conversely enhanced protein stability and DNA-binding affinity. The mNANOG L122A, when overexpressed in mouse embryonic stem cells, maintained their expression of self-renewal markers even when retinoic acid was added to forcibly drive differentiation. When overexpressed in epiblast stem cells or human induced pluripotent stem cells, the L122A mutants enhanced reprogramming into ground-state pluripotency. These findings demonstrate that structural and biophysical information on key transcriptional factors provides insights into the manipulation of stem cell behaviors and a framework for rational protein engineering.


Assuntos
Proliferação de Células/genética , Reprogramação Celular/genética , Proteínas de Homeodomínio/genética , Mutação , Células-Tronco Pluripotentes/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , Células Cultivadas , Cristalografia por Raios X , DNA/química , DNA/genética , DNA/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos Endogâmicos C57BL , Modelos Moleculares , Dados de Sequência Molecular , Proteína Homeobox Nanog , Conformação de Ácido Nucleico , Células-Tronco Pluripotentes/citologia , Regiões Promotoras Genéticas/genética , Ligação Proteica , Estrutura Terciária de Proteína , Transfecção
10.
Proc Natl Acad Sci U S A ; 112(8): 2467-72, 2015 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-25675535

RESUMO

Colorectal cancers (CRCs) account for nearly 10% of all cancer deaths in industrialized countries. Recent evidence points to a central role for the nuclear receptor liver receptor homolog-1 (LRH-1) in intestinal tumorigenesis. Interaction of LRH-1 with the Wnt/ß-catenin pathway, highly active in a critical subpopulation of CRC cells, underscores the importance of elucidating LRH-1's role in this disease. Reduction of LRH-1 diminishes tumor burden in murine models of CRC; however, it is not known whether LRH-1 is required for tumorigenesis, for proliferation, or for both. In this work, we address this question through shRNA-mediated silencing of LRH-1 in established CRC cell lines. LRH-1 mRNA knockdown results in significantly impaired proliferation in a cell line highly expressing the receptor and more modest impairment in a cell line with moderate LRH-1 expression. Cell-cycle analysis shows prolongation of G0/G1 with LRH-1 silencing, consistent with LRH-1 cell-cycle influences in other tissues. Cluster analysis of microarray gene expression demonstrates significant genome wide alterations with major effects in cell-cycle regulation, signal transduction, bile acid and cholesterol metabolism, and control of apoptosis. This study demonstrates a critical proproliferative role for LRH-1 in established colon cancer cell lines. LRH-1 exerts its effects via multiple signaling networks. Our results suggest that selected CRC patients could benefit from LRH-1 inhibitors.


Assuntos
Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Receptores Citoplasmáticos e Nucleares/genética , Células CACO-2 , Ciclo Celular/genética , Proliferação de Células , Técnicas de Silenciamento de Genes , Células HT29 , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Citoplasmáticos e Nucleares/metabolismo , Reprodutibilidade dos Testes
11.
Nat Commun ; 6: 6204, 2015 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-25686748

RESUMO

The retinoblastoma binding protein KDM5A removes methyl marks from lysine 4 of histone H3 (H3K4). Misregulation of KDM5A contributes to the pathogenesis of lung and gastric cancers. In addition to its catalytic jumonji C domain, KDM5A contains three PHD reader domains, commonly recognized as chromatin recruitment modules. It is unknown whether any of these domains in KDM5A have functions beyond recruitment and whether they regulate the catalytic activity of the demethylase. Here using biochemical and nuclear magnetic resonance (NMR)-based structural studies, we show that the PHD1 preferentially recognizes unmethylated H3K4 histone tail, product of KDM5A-mediated demethylation of tri-methylated H3K4 (H3K4me3). Binding of unmodified H3 peptide to the PHD1 stimulates catalytic domain-mediated removal of methyl marks from H3K4me3 peptide and nucleosome substrates. This positive-feedback mechanism--enabled by the functional coupling between a reader and a catalytic domain in KDM5A--suggests a model for the spread of demethylation on chromatin.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteína 2 de Ligação ao Retinoblastoma/metabolismo , Algoritmos , Sequência de Aminoácidos , Animais , Domínio Catalítico , Linhagem Celular , Cromatina/química , Dicroísmo Circular , Glutationa Transferase/metabolismo , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/química , Insetos , Cinética , Lisina/química , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Nucleossomos/química , Peptídeos/química , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos
12.
Trends Biochem Sci ; 40(1): 25-35, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25499868

RESUMO

Nuclear receptors (NRs) are key players in the regulation of gene expression, coordinating protein assemblies upon their surfaces. NRs are regulated by ligand binding, which remodels the interaction surfaces and subsequently influences macromolecular complex formation. Structural biology has been instrumental in the discovery of some of these ligands, but there are still orphan NRs (ONRs) whose bona fide ligands have yet to be identified. Over the past decade, fundamental structural and functional breakthroughs have led to a deeper understanding of ONR actions and their multidomain organization. Here, we summarize the structural advances in ONRs with implications for the therapeutic treatment of diseases such as metabolic syndrome and cancer.


Assuntos
Proteínas de Ligação a DNA/química , Receptores Nucleares Órfãos/química , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais , Sítios de Ligação , Cristalografia por Raios X , Proteínas de Ligação a DNA/metabolismo , Humanos , Ligantes , Receptores Nucleares Órfãos/metabolismo , Conformação Proteica , Relação Estrutura-Atividade
13.
Proc Natl Acad Sci U S A ; 111(42): 15054-9, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25288771

RESUMO

The signaling phosphatidylinositol lipids PI(4,5)P2 (PIP2) and PI(3,4,5)P3 (PIP3) bind nuclear receptor 5A family (NR5As), but their regulatory mechanisms remain unknown. Here, the crystal structures of human NR5A1 (steroidogenic factor-1, SF-1) ligand binding domain (LBD) bound to PIP2 and PIP3 show the lipid hydrophobic tails sequestered in the hormone pocket, as predicted. However, unlike classic nuclear receptor hormones, the phosphoinositide head groups are fully solvent-exposed and complete the LBD fold by organizing the receptor architecture at the hormone pocket entrance. The highest affinity phosphoinositide ligand PIP3 stabilizes the coactivator binding groove and increases coactivator peptide recruitment. This receptor-ligand topology defines a previously unidentified regulatory protein-lipid surface on SF-1 with the phosphoinositide head group at its nexus and poised to interact with other proteins. This surface on SF-1 coincides with the predicted binding site of the corepressor DAX-1 (dosage-sensitive sex reversal, adrenal hypoplasia critical region on chromosome X), and importantly harbors missense mutations associated with human endocrine disorders. Our data provide the structural basis for this poorly understood cluster of human SF-1 mutations and demonstrates how signaling phosphoinositides function as regulatory ligands for NR5As.


Assuntos
Fosfatidilinositóis/química , Fator Esteroidogênico 1/química , Aminoácidos/química , Animais , Transporte Biológico , Núcleo Celular/metabolismo , Cromatografia , Simulação por Computador , Cristalografia por Raios X , Elétrons , Humanos , Ligantes , Lipídeos/química , Camundongos , Modelos Moleculares , Conformação Molecular , Mutação , Mutação de Sentido Incorreto , Peptídeos/química , Transdução de Sinais , Solventes/química , Ressonância de Plasmônio de Superfície , Propriedades de Superfície , Temperatura , Água/química
15.
PLoS One ; 9(6): e99440, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24936658

RESUMO

Nuclear receptors (NRs) are an important group of ligand-dependent transcriptional factors. Presently, no natural or synthetic ligand has been identified for a large group of orphan NRs. Small molecules to target these orphan NRs will provide unique resources for uncovering regulatory systems that impact human health and to modulate these pathways with drugs. The orphan NR tailless (TLX, NR2E1), a transcriptional repressor, is a major player in neurogenesis and Neural Stem Cell (NSC) derived brain tumors. No chemical probes that modulate TLX activity are available, and it is not clear whether TLX is druggable. To assess TLX ligand binding capacity, we created homology models of the TLX ligand binding domain (LBD). Results suggest that TLX belongs to an emerging class of NRs that lack LBD helices α1 and α2 and that it has potential to form a large open ligand binding pocket (LBP). Using a medium throughput screening strategy, we investigated direct binding of 20,000 compounds to purified human TLX protein and verified interactions with a secondary (orthogonal) assay. We then assessed effects of verified binders on TLX activity using luciferase assays. As a result, we report identification of three compounds (ccrp1, ccrp2 and ccrp3) that bind to recombinant TLX protein with affinities in the high nanomolar to low micromolar range and enhance TLX transcriptional repressive activity. We conclude that TLX is druggable and propose that our lead compounds could serve as scaffolds to derive more potent ligands. While our ligands potentiate TLX repressive activity, the question of whether it is possible to develop ligands to de-repress TLX activity remains open.


Assuntos
Didrogesterona/farmacologia , Piperazinas/farmacologia , Pirazóis/farmacologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Ativação Transcricional/efeitos dos fármacos , Sequência de Aminoácidos , Sítios de Ligação , Fator II de Transcrição COUP/antagonistas & inibidores , Fator II de Transcrição COUP/fisiologia , Receptor beta de Estrogênio/antagonistas & inibidores , Receptor beta de Estrogênio/fisiologia , Genes Reporter , Células HeLa , Humanos , Concentração Inibidora 50 , Ligantes , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Modelos Moleculares , Dados de Sequência Molecular , Receptores Nucleares Órfãos , Ligação Proteica , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/química , Receptor X Retinoide alfa/antagonistas & inibidores , Receptor X Retinoide alfa/fisiologia , Transcrição Gênica/efeitos dos fármacos
16.
Proc Natl Acad Sci U S A ; 111(25): 9103-8, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24927529

RESUMO

Transcription factor II D (TFIID) is a multiprotein complex that nucleates formation of the basal transcription machinery. TATA binding protein-associated factors 1 and 7 (TAF1 and TAF7), two subunits of TFIID, are integral to the regulation of eukaryotic transcription initiation and play key roles in preinitiation complex (PIC) assembly. Current models suggest that TAF7 acts as a dissociable inhibitor of TAF1 histone acetyltransferase activity and that this event ensures appropriate assembly of the RNA polymerase II-mediated PIC before transcriptional initiation. Here, we report the 3D structure of a complex of yeast TAF1 with TAF7 at 2.9 Å resolution. The structure displays novel architecture and is characterized by a large predominantly hydrophobic heterodimer interface and extensive cofolding of TAF subunits. There are no obvious similarities between TAF1 and known histone acetyltransferases. Instead, the surface of the TAF1-TAF7 complex contains two prominent conserved surface pockets, one of which binds selectively to an inhibitory trimethylated histone H3 mark on Lys27 in a manner that is also regulated by phosphorylation at the neighboring H3 serine. Our findings could point toward novel roles for the TAF1-TAF7 complex in regulation of PIC assembly via reading epigenetic histone marks.


Assuntos
Histona Acetiltransferases/química , Complexos Multiproteicos/química , Fatores Associados à Proteína de Ligação a TATA/química , Fator de Transcrição TFIID/química , Sítios de Ligação , Histonas/química , Humanos , Ligação Proteica , Estrutura Quaternária de Proteína
17.
PLoS One ; 8(6): e66669, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23805258

RESUMO

Kinesin-like calmodulin binding protein (KCBP), a Kinesin-14 family motor protein, is involved in the structural organization of microtubules during mitosis and trichome morphogenesis in plants. The molecular mechanism of microtubule bundling by KCBP remains unknown. KCBP binding to microtubules is regulated by Ca(2+)-binding proteins that recognize its C-terminal regulatory domain. In this work, we have discovered a new function of the regulatory domain. We present a crystal structure of an Arabidopsis KCBP fragment showing that the C-terminal regulatory domain forms a dimerization interface for KCBP. This dimerization site is distinct from the dimerization interface within the N-terminal domain. Side chains of hydrophobic residues of the calmodulin binding helix of the regulatory domain form the C-terminal dimerization interface. Biochemical experiments show that another segment of the regulatory domain located beyond the dimerization interface, its negatively charged coil, is unexpectedly and absolutely required to stabilize the dimers. The strong microtubule bundling properties of KCBP are unaffected by deletion of the C-terminal regulatory domain. The slow minus-end directed motility of KCBP is also unchanged in vitro. Although the C-terminal domain is not essential for microtubule bundling, we suggest that KCBP may use its two independent dimerization interfaces to support different types of bundled microtubule structures in cells. Two distinct dimerization sites may provide a mechanism for microtubule rearrangement in response to Ca(2+) signaling since Ca(2+)- binding proteins can disengage KCBP dimers dependent on its C-terminal dimerization interface.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Cinesinas/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Proteínas de Ligação a Calmodulina/química , Proteínas de Ligação a Calmodulina/genética , Cristalografia por Raios X , Dimerização , Cinesinas/química , Microscopia de Interferência , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Ultracentrifugação
18.
J Biol Chem ; 288(27): 19830-44, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23667258

RESUMO

Liver receptor homolog 1 (nuclear receptor LRH-1, NR5A2) is an essential regulator of gene transcription, critical for maintenance of cell pluripotency in early development and imperative for the proper functions of the liver, pancreas, and intestines during the adult life. Although physiological hormones of LRH-1 have not yet been identified, crystallographic and biochemical studies demonstrated that LRH-1 could bind regulatory ligands and suggested phosphatidylinositols as potential hormone candidates for this receptor. No synthetic antagonists of LRH-1 are known to date. Here, we identify the first small molecule antagonists of LRH-1 activity. Our search for LRH-1 modulators was empowered by screening of 5.2 million commercially available compounds via molecular docking followed by verification of the top-ranked molecules using in vitro direct binding and transcriptional assays. Experimental evaluation of the predicted ligands identified two compounds that inhibit the transcriptional activity of LRH-1 and diminish the expression of the receptor's target genes. Among the affected transcriptional targets are co-repressor SHP (small heterodimer partner) as well as cyclin E1 (CCNE1) and G0S2 genes that are known to regulate cell growth and proliferation. Treatments of human pancreatic (AsPC-1), colon (HT29), and breast adenocarcinoma cells T47D and MDA-MB-468 with the LRH-1 antagonists resulted in the receptor-mediated inhibition of cancer cell proliferation. Our data suggest that specific antagonists of LRH-1 could be used as specific molecular probes for elucidating the roles of the receptor in different types of malignancies.


Assuntos
Proliferação de Células/efeitos dos fármacos , Descoberta de Drogas , Sondas Moleculares , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Ciclina E/química , Ciclina E/genética , Ciclina E/metabolismo , Células HEK293 , Células HeLa , Humanos , Sondas Moleculares/química , Sondas Moleculares/farmacologia , Neoplasias/química , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Proteínas Oncogênicas/química , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Receptores Citoplasmáticos e Nucleares/química , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Relação Estrutura-Atividade
19.
PLoS One ; 7(9): e43761, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22984442

RESUMO

The human nuclear factor related to kappa-B-binding protein (NFRKB) is a 1299-residue protein that is a component of the metazoan INO80 complex involved in chromatin remodeling, transcription regulation, DNA replication and DNA repair. Although full length NFRKB is predicted to be around 65% disordered, comparative sequence analysis identified several potentially structured sections in the N-terminal region of the protein. These regions were targeted for crystallographic studies, and the structure of one of these regions spanning residues 370-495 was determined using the JCSG high-throughput structure determination pipeline. The structure reveals a novel, mostly helical domain reminiscent of the winged-helix fold typically involved in DNA binding. However, further analysis shows that this domain does not bind DNA, suggesting it may belong to a small group of winged-helix domains involved in protein-protein interactions.


Assuntos
Proteínas de Ligação a DNA/química , Fatores de Transcrição Winged-Helix/química , Sequência de Aminoácidos , Proteínas de Bactérias/química , Cristalografia por Raios X , Proteínas Culina/química , DNA/metabolismo , Humanos , Dados de Sequência Molecular , Ligação Proteica , Desnaturação Proteica , Mapas de Interação de Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Repressoras/química , Saccharomyces cerevisiae/química , Alinhamento de Sequência , Homologia Estrutural de Proteína , Temperatura
20.
Mol Cell Endocrinol ; 348(2): 394-402, 2012 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-21878368

RESUMO

Nuclear receptors (NRs) form a large superfamily of transcription factors that participate in virtually every key biological process. They control development, fertility, gametogenesis and are misregulated in many cancers. Their enormous functional plasticity as transcription factors relates in part to NR-mediated interactions with hundreds of coregulatory proteins upon ligand (e.g., hormone) binding to their ligand binding domains (LBD), or following covalent modification. Some coregulator association relates to the distinct residues that shape a coactivator binding pocket termed AF-2, a surface groove that primarily determines the preference and specificity of protein-protein interactions. However, the highly conserved AF-2 pocket in the NR superfamily appears to be insufficient to account for NR subtype specificity leading to fine transcriptional modulation in certain settings. Additional protein-protein interaction surfaces, most notably on their LBD, may contribute to modulating NR function. NR coregulators and chaperones, normally much larger than the NR itself, may also bind to such interfaces. In the case of the androgen receptor (AR) LBD surface, structural and functional data highlighted the presence of another site named BF-3, which lies at a distinct but topographically adjacent surface to AF-2. AR BF-3 is a hot spot for mutations involved in prostate cancer and androgen insensitivity syndromes, and some FDA-approved drugs bind at this site. Structural studies suggested an allosteric relationship between AF-2 and BF-3, as occupancy of the latter affected coactivator recruitment to AF-2. Physiological relevant partners of AR BF-3 have not been described as yet. The newly discovered site is highly conserved among the steroid receptors subclass, but is also present in other NRs. Several missense mutations in the BF-3 regions of these human NRs are implicated in pathology and affect their function in vitro. The fact that AR BF-3 pocket is a druggable site evidences its pharmacological potential. Compounds that may affect allosterically NR function by binding to BF-3 open promising avenues to develop type-specific NR modulators.


Assuntos
Receptores Citoplasmáticos e Nucleares/química , Regulação Alostérica , Motivos de Aminoácidos/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação/genética , Sequência Conservada , Humanos , Mutação , Estrutura Terciária de Proteína , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/fisiologia , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...