Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 299(12): 105475, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37981208

RESUMEN

Heterozygous GRN (progranulin) mutations cause frontotemporal dementia (FTD) due to haploinsufficiency, and increasing progranulin levels is a major therapeutic goal. Several microRNAs, including miR-29b, negatively regulate progranulin protein levels. Antisense oligonucleotides (ASOs) are emerging as a promising therapeutic modality for neurological diseases, but strategies for increasing target protein levels are limited. Here, we tested the efficacy of ASOs as enhancers of progranulin expression by sterically blocking the miR-29b binding site in the 3' UTR of the human GRN mRNA. We found 16 ASOs that increase progranulin protein in a dose-dependent manner in neuroglioma cells. A subset of these ASOs also increased progranulin protein in iPSC-derived neurons and in a humanized GRN mouse model. In FRET-based assays, the ASOs effectively competed for miR-29b from binding to the GRN 3' UTR RNA. The ASOs increased levels of newly synthesized progranulin protein by increasing its translation, as revealed by polysome profiling. Together, our results demonstrate that ASOs can be used to effectively increase target protein levels by partially blocking miR binding sites. This ASO strategy may be therapeutically feasible for progranulin-deficient FTD as well as other conditions of haploinsufficiency.


Asunto(s)
Demencia Frontotemporal , MicroARNs , Oligonucleótidos Antisentido , Progranulinas , Animales , Humanos , Ratones , Regiones no Traducidas 3' , Sitios de Unión , Demencia Frontotemporal/genética , Péptidos y Proteínas de Señalización Intercelular/genética , MicroARNs/genética , Mutación , Oligonucleótidos Antisentido/genética , Progranulinas/genética , ARN Mensajero/genética
2.
Pharmacol Rev ; 75(6): 1233-1318, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37586884

RESUMEN

The NR superfamily comprises 48 transcription factors in humans that control a plethora of gene network programs involved in a wide range of physiologic processes. This review will summarize and discuss recent progress in NR biology and drug development derived from integrating various approaches, including biophysical techniques, structural studies, and translational investigation. We also highlight how defective NR signaling results in various diseases and disorders and how NRs can be targeted for therapeutic intervention via modulation via binding to synthetic lipophilic ligands. Furthermore, we also review recent studies that improved our understanding of NR structure and signaling. SIGNIFICANCE STATEMENT: Nuclear receptors (NRs) are ligand-regulated transcription factors that are critical regulators of myriad physiological processes. NRs serve as receptors for an array of drugs, and in this review, we provide an update on recent research into the roles of these drug targets.


Asunto(s)
Farmacología Clínica , Humanos , Receptores Citoplasmáticos y Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Portadoras , Ligandos
3.
Eur J Med Chem ; 258: 115582, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37421886

RESUMEN

Estrogen-related receptors (ERR) are an orphan nuclear receptor sub-family that play a critical role in regulating gene transcription for several physiological processes including mitochondrial function, cellular energy utilization and homeostasis. They have also been implicated to play a role in several pathological conditions. Herein, we report the identification, synthesis, structure-activity relationships and pharmacological evaluation of a new chemical series of potent pan-ERR agonists. This template was designed for ERRγ starting from the known acyl hydrazide template and compounds such as agonist GSK-4716 employing a structure-based drug design approach. This led to the preparation of a series of 2,5-disubstituted thiophenes from which several were found to be potent agonists of ERRγ in cell-based co-transfection assays. Additionally, direct binding to ERRγ was established through 1H NMR protein-ligand binding experiments. Compound optimization revealed that the phenolic or aniline groups could be replaced with a boronic acid moiety, which was able to maintain activity and demonstrated improved metabolic stability in microsomal in vitro assays. Further pharmacological evaluation of these compounds showed that they had roughly equivalent agonist activity on ERR isoforms α and ß representing an ERR pan-agonist profile. One potent agonist, SLU-PP-915 (10s), which contained a boronic acid moiety was profiled in gene expression assays and found to significantly upregulate the expression of ERR target genes such as peroxisome-proliferator activated receptor γ co-activators-1α, lactate dehydrogenase A, DNA damage inducible transcript 4 and pyruvate dehydrogenase kinase 4 both in vitro and in vivo.


Asunto(s)
Estrógenos , Isoformas de Proteínas
4.
SLAS Discov ; 27(1): 8-19, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35058179

RESUMEN

The severe acute respiratory syndrome coronavirus 2 responsible for COVID-19 remains a persistent threat to mankind, especially for the immunocompromised and elderly for which the vaccine may have limited effectiveness. Entry of SARS-CoV-2 requires a high affinity interaction of the viral spike protein with the cellular receptor angiotensin-converting enzyme 2. Novel mutations on the spike protein correlate with the high transmissibility of new variants of SARS-CoV-2, highlighting the need for small molecule inhibitors of virus entry into target cells. We report the identification of such inhibitors through a robust high-throughput screen testing 15,000 small molecules from unique libraries. Several leads were validated in a suite of mechanistic assays, including whole cell SARS-CoV-2 infectivity assays. The main lead compound, calpeptin, was further characterized using SARS-CoV-1 and the novel SARS-CoV-2 variant entry assays, SARS-CoV-2 protease assays and molecular docking. This study reveals calpeptin as a potent and specific inhibitor of SARS-CoV-2 and some variants.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Inhibidores de Cisteína Proteinasa/farmacología , Dipéptidos/farmacología , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Catepsina L/antagonistas & inhibidores , Línea Celular , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Células HEK293 , Humanos , Simulación del Acoplamiento Molecular , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/crecimiento & desarrollo , Serina Endopeptidasas/metabolismo , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células Vero
6.
J Med Chem ; 63(24): 15639-15654, 2020 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-33289551

RESUMEN

Nurr1/NR4A2 is an orphan nuclear receptor transcription factor implicated as a drug target for neurological disorders including Alzheimer's and Parkinson's diseases. Previous studies identified small-molecule NR4A nuclear receptor modulators, but it remains unclear if these ligands affect transcription via direct binding to Nurr1. We assessed 12 ligands reported to affect NR4A activity for Nurr1-dependent and Nurr1-independent transcriptional effects and the ability to bind the Nurr1 ligand-binding domain (LBD). Protein NMR structural footprinting data show that amodiaquine, chloroquine, and cytosporone B bind the Nurr1 LBD; ligands that do not bind include C-DIM12, celastrol, camptothecin, IP7e, isoalantolactone, ethyl 2-[2,3,4-trimethoxy-6-(1-octanoyl)phenyl]acetate (TMPA), and three high-throughput screening hit derivatives. Importantly, ligands that modulate Nurr1 transcription also show Nurr1-independent effects on transcription in a cell type-specific manner, indicating that care should be taken when interpreting the functional response of these ligands in transcriptional assays. These findings should help focus medicinal chemistry efforts that desire to optimize Nurr1-binding ligands.


Asunto(s)
Ligandos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Amodiaquina/química , Amodiaquina/metabolismo , Amodiaquina/farmacología , Animales , Línea Celular , Cloroquina/química , Cloroquina/metabolismo , Cloroquina/farmacología , Humanos , Resonancia Magnética Nuclear Biomolecular , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/química , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Fenilacetatos/química , Fenilacetatos/metabolismo , Fenilacetatos/farmacología , Unión Proteica , Ratas , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Transcripción Genética/efectos de los fármacos
7.
Cell Chem Biol ; 27(10): 1272-1284.e4, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32763139

RESUMEN

TLX is an orphan nuclear receptor that plays a critical role in both embryonic and adult neurogenesis, as well in the pathogenesis of glioblastomas. TLX functions predominately as a transcriptional repressor, but no natural ligands or high-affinity synthetic ligands have been identified. Here, we describe the identification of natural and synthetic retinoids as functional ligands for TLX. We identified potent synthetic retinoids that directly bind to TLX and either activate or inhibit its transcriptional repressor activity. Furthermore, we identified all-trans and 11-cis retinaldehyde (retinal), retinoids that play an essential role in the visual cycle, as the preferential natural retinoids that bind to and modulate the function of TLX. Molecular dynamics simulations followed by mutational analysis provided insight into the molecular basis of retinoid binding to TLX. Our data support the validity of TLX as a target for development of therapeutics to treat cognitive disorders and/or glioblastomas.


Asunto(s)
Productos Biológicos/química , Receptores Citoplasmáticos y Nucleares/química , Retinoides/química , Sitios de Unión/efectos de los fármacos , Productos Biológicos/síntesis química , Productos Biológicos/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Masculino , Simulación de Dinámica Molecular , Estructura Molecular , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/agonistas , Retinoides/síntesis química , Retinoides/farmacología , Adulto Joven
8.
FASEB J ; 33(7): 8280-8293, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31021670

RESUMEN

The HIV-1 transactivation protein (Tat) binds the HIV mRNA transactivation responsive element (TAR), regulating transcription and reactivation from latency. Drugs against Tat are unfortunately not clinically available. We reported that didehydro-cortistatin A (dCA) inhibits HIV-1 Tat activity. In human CD4+ T cells isolated from aviremic individuals and in the humanized mouse model of latency, combining dCA with antiretroviral therapy accelerates HIV-1 suppression and delays viral rebound upon treatment interruption. This drug class is amenable to block-and-lock functional cure approaches, aimed at a durable state of latency. Simian immunodeficiency virus (SIV) infection of rhesus macaques (RhMs) is the best-characterized model for AIDS research. Here, we demonstrate, using in vitro and cell-based assays, that dCA directly binds to SIV Tat's basic domain. dCA specifically inhibits SIV Tat binding to TAR, but not a Tat-Rev fusion protein, which activates transcription when Rev binds to its cognate RNA binding site replacing the apical region of TAR. Tat-TAR inhibition results in loss of RNA polymerase II recruitment to the SIV promoter. Importantly, dCA potently inhibits SIV reactivation from latently infected Hut78 cells and from primary CD4+ T cells explanted from SIVmac239-infected RhMs. In sum, dCA's remarkable breadth of activity encourages SIV-infected RhM use for dCA preclinical evaluation.-Mediouni, S., Kessing, C. F., Jablonski, J. A., Thenin-Houssier, S., Clementz, M., Kovach, M. D., Mousseau, G., de Vera, I.M.S., Li, C., Kojetin, D. J., Evans, D. T., Valente, S. T. The Tat inhibitor didehydro-cortistatin A suppresses SIV replication and reactivation.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Productos del Gen tat/antagonistas & inhibidores , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Virus de la Inmunodeficiencia de los Simios/fisiología , Activación Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Productos del Gen tat/metabolismo , Células HEK293 , Células HeLa , Compuestos Heterocíclicos de 4 o más Anillos , Humanos , Isoquinolinas , Macaca mulatta , Regiones Promotoras Genéticas , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Secuencias Repetidas Terminales
9.
mBio ; 10(1)2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30723126

RESUMEN

The intrinsically disordered HIV-1 Tat protein binds the viral RNA transactivation response structure (TAR), which recruits transcriptional cofactors, amplifying viral mRNA expression. Limited Tat transactivation correlates with HIV-1 latency. Unfortunately, Tat inhibitors are not clinically available. The small molecule didehydro-cortistatin A (dCA) inhibits Tat, locking HIV-1 in persistent latency, blocking viral rebound. We generated chemical derivatives of dCA that rationalized molecular docking of dCA to an active and specific Tat conformer. These revealed the importance of the cycloheptene ring and the isoquinoline nitrogen's positioning in the interaction with specific residues of Tat's basic domain. These features are distinct from the ones required for inhibition of cyclin-dependent kinase 8 (CDK8), the only other known ligand of dCA. Besides, we demonstrated that dCA activity on HIV-1 transcription is independent of CDK8. The binding of dCA to Tat with nanomolar affinity alters the local protein environment, rendering Tat more resistant to proteolytic digestion. dCA thus locks a transient conformer of Tat, specifically blocking functions dependent of its basic domain, namely the Tat-TAR interaction; while proteins with similar basic patches are unaffected by dCA. Our results improve our knowledge of the mode of action of dCA and support structure-based design strategies targeting Tat, to help advance development of dCA, as well as novel Tat inhibitors.IMPORTANCE Tat activates virus production, and limited Tat transactivation correlates with HIV-1 latency. The Tat inhibitor dCA locks HIV in persistent latency. This drug class enables block-and-lock functional cure approaches, aimed at reducing residual viremia during therapy and limiting viral rebound. dCA may also have additional therapeutic benefits since Tat is also neurotoxic. Unfortunately, Tat inhibitors are not clinically available. We generated chemical derivatives and rationalized binding to an active and specific Tat conformer. dCA features required for Tat inhibition are distinct from features needed for inhibition of cyclin-dependent kinase 8 (CDK8), the only other known target of dCA. Furthermore, knockdown of CDK8 did not impact dCA's activity on HIV-1 transcription. Binding of dCA to Tat's basic domain altered the local protein environment and rendered Tat more resistant to proteolytic digestion. dCA locks a transient conformer of Tat, blocking functions dependent on its basic domain, namely its ability to amplify viral transcription. Our results define dCA's mode of action, support structure-based-design strategies targeting Tat, and provide valuable information for drug development around the dCA pharmacophore.


Asunto(s)
Fármacos Anti-VIH/metabolismo , VIH-1/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/metabolismo , Isoquinolinas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Fármacos Anti-VIH/síntesis química , Quinasa 8 Dependiente de Ciclina/metabolismo , Células HeLa , Compuestos Heterocíclicos de 4 o más Anillos/síntesis química , Humanos , Isoquinolinas/síntesis química , Simulación del Acoplamiento Molecular , Unión Proteica
10.
Structure ; 27(1): 66-77.e5, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30416039

RESUMEN

Nuclear receptor-related 1 protein (Nurr1/NR4A2) is an orphan nuclear receptor (NR) that is considered to function without a canonical ligand-binding pocket (LBP). A crystal structure of the Nurr1 ligand-binding domain (LBD) revealed no physical space in the conserved region where other NRs with solvent accessible apo-protein LBPs bind synthetic and natural ligands. Using solution nuclear magnetic resonance spectroscopy, hydrogen/deuterium exchange mass spectrometry, and molecular dynamics simulations, we show that the putative canonical Nurr1 LBP is dynamic with high solvent accessibility, exchanges between two or more conformations on the microsecond-to-millisecond timescale, and can expand from the collapsed crystallized conformation to allow binding of unsaturated fatty acids. These findings should stimulate future studies to probe the ligandability and druggability of Nurr1 for both endogenous and synthetic ligands, which could lead to new therapeutics for Nurr1-related diseases, including Parkinson's disease and schizophrenia.


Asunto(s)
Simulación del Acoplamiento Molecular , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/química , Sitios de Unión , Ácidos Grasos Insaturados/química , Humanos , Ligandos , Simulación de Dinámica Molecular , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Unión Proteica
11.
Nat Commun ; 9(1): 1794, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29728618

RESUMEN

The nuclear receptor ligand-binding domain (LBD) is a highly dynamic entity. Crystal structures have defined multiple low-energy LBD structural conformations of the activation function-2 (AF-2) co-regulator-binding surface, yet it remains unclear how ligand binding influences the number and population of conformations within the AF-2 structural ensemble. Here, we present a nuclear receptor co-regulator-binding surface structural ensemble in solution, viewed through the lens of fluorine-19 (19F) nuclear magnetic resonance (NMR) and molecular simulations, and the response of this ensemble to ligands, co-regulator peptides and heterodimerization. We correlate the composition of this ensemble with function in peroxisome proliferator-activated receptor-γ (PPARγ) utilizing ligands of diverse efficacy in co-regulator recruitment. While the co-regulator surface of apo PPARγ and partial-agonist-bound PPARγ is characterized by multiple thermodynamically accessible conformations, the full and inverse-agonist-bound PPARγ co-regulator surface is restricted to a few conformations which favor coactivator or corepressor binding, respectively.


Asunto(s)
Simulación de Dinámica Molecular , PPAR gamma/química , Péptidos/química , Conformación Proteica , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Ligandos , Espectroscopía de Resonancia Magnética , PPAR gamma/agonistas , PPAR gamma/metabolismo , Péptidos/metabolismo , Unión Proteica , Multimerización de Proteína , Termodinámica
12.
ACS Pharmacol Transl Sci ; 1(2): 134-137, 2018 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-32219209

RESUMEN

Over the past decade, advances in biophysical chemistry, genomic analysis, and structural biology have resulted in the exponential growth of knowledge and critical insight into the function and regulation of orphan nuclear receptors. This article summarizes the current progress in illuminating the structure, function, and regulation of orphan nuclear receptors and their involvement in the physiology, development and molecular mechanism of different pathological conditions. Moreover, current strategies for discovering endogenous ligands, downstream NR-regulated target genes, and new drugs for future therapeutics will be discussed.

13.
Structure ; 25(10): 1506-1518.e4, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28890360

RESUMEN

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.


Asunto(s)
ADN/metabolismo , Complejos Multiproteicos/química , Coactivador 2 del Receptor Nuclear/química , Coactivador 2 del Receptor Nuclear/metabolismo , Sitios de Unión , Espectroscopía de Resonancia Magnética con Carbono-13 , Medición de Intercambio de Deuterio , Regulación de la Expresión Génica , Humanos , Ligandos , PPAR gamma/química , PPAR gamma/metabolismo , Unión Proteica , Receptor alfa X Retinoide/química , Receptor alfa X Retinoide/metabolismo
14.
Nucleic Acids Res ; 45(14): 8596-8608, 2017 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-28591827

RESUMEN

The glucocorticoid receptor (GR) is a ligand-regulated transcription factor that controls the expression of extensive gene networks, driving both up- and down-regulation. GR utilizes multiple DNA-binding-dependent and -independent mechanisms to achieve context-specific transcriptional outcomes. The DNA-binding-independent mechanism involves tethering of GR to the pro-inflammatory transcription factor activator protein-1 (AP-1) through protein-protein interactions. This mechanism has served as the predominant model of GR-mediated transrepression of inflammatory genes. However, ChIP-seq data have consistently shown GR to occupy AP-1 response elements (TREs), even in the absence of AP-1. Therefore, the current model is insufficient to explain GR action at these sites. Here, we show that GR regulates a subset of inflammatory genes in a DNA-binding-dependent manner. Using structural biology and biochemical approaches, we show that GR binds directly to TREs via sequence-specific contacts to a GR-binding sequence (GBS) half-site found embedded within the TRE motif. Furthermore, we show that GR-mediated transrepression observed at TRE sites to be DNA-binding-dependent. This represents a paradigm shift in the field, showing that GR uses multiple mechanisms to suppress inflammatory gene expression. This work further expands our understanding of this complex multifaceted transcription factor.


Asunto(s)
Regulación de la Expresión Génica , Inflamación/genética , Receptores de Glucocorticoides/genética , Elementos de Respuesta/genética , Factor de Transcripción AP-1/genética , Secuencia de Bases , Sitios de Unión/genética , Línea Celular Tumoral , Cristalografía por Rayos X , ADN/química , ADN/genética , ADN/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/metabolismo , Factor de Transcripción AP-1/química , Factor de Transcripción AP-1/metabolismo
15.
J Med Chem ; 59(22): 10335-10341, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27783520

RESUMEN

In a previous study, a cocrystal structure of PPARγ bound to 2-chloro-N-(3-chloro-4-((5-chlorobenzo[d]thiazol-2-yl)thio)phenyl)-4-(trifluoromethyl)benzenesulfonamide (1, T2384) revealed two orthosteric pocket binding modes attributed to a concentration-dependent biochemical activity profile. However, 1 also bound an alternate/allosteric site that could alternatively account for the profile. Here, we show ligand aggregation afflicts the activity profile of 1 in biochemical assays. However, ligand-observed fluorine (19F) and protein-observed NMR confirms 1 binds PPARγ with two orthosteric binding modes and to an allosteric site.


Asunto(s)
Sitio Alostérico/efectos de los fármacos , Benzotiazoles/farmacología , PPAR gamma/agonistas , Sulfonamidas/farmacología , Benzotiazoles/química , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Estructura Molecular , Relación Estructura-Actividad , Sulfonamidas/química
16.
FEBS J ; 283(19): 3567-3586, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27459543

RESUMEN

Here we provide the first detailed biochemical study of a noncanonical E1-like enzyme with broad specificity for cognate ubiquitin-like (Ubl) proteins that mediates Ubl protein modification and sulfur mobilization to form molybdopterin and thiolated tRNA. Isothermal titration calorimetry and in vivo analyses proved useful in discovering that environmental conditions, ATP binding, and Ubl type controlled the mechanism of association of the Ubl protein with its cognate E1-like enzyme (SAMP and UbaA of the archaeon Haloferax volcanii, respectively). Further analysis revealed that ATP hydrolysis triggered the formation of thioester and peptide bonds within the Ubl:E1-like complex. Importantly, the thioester was an apparent precursor to Ubl protein modification but not sulfur mobilization. Comparative modeling to MoeB/ThiF guided the discovery of key residues within the adenylation domain of UbaA that were needed to bind ATP as well as residues that were specifically needed to catalyze the downstream reactions of sulfur mobilization and/or Ubl protein modification. UbaA was also found to be Ubl-automodified at lysine residues required for early (ATP binding) and late (sulfur mobilization) stages of enzyme activity revealing multiple layers of autoregulation. Cysteine residues, distinct from the canonical E1 'active site' cysteine, were found important in UbaA function supporting a model that this noncanonical E1 is structurally flexible in its active site to allow Ubl~adenylate, Ubl~E1-like thioester and cysteine persulfide(s) intermediates to form.


Asunto(s)
Proteínas Arqueales/química , Proteínas Arqueales/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Azufre/metabolismo , Enzimas Activadoras de Ubiquitina/química , Enzimas Activadoras de Ubiquitina/metabolismo , Adenosina Trifosfato/metabolismo , Cisteína/fisiología , Haloferax volcanii/enzimología , Ligandos , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/química , Compuestos de Sulfhidrilo/metabolismo , Termodinámica , Ubiquitinación
17.
ACS Chem Biol ; 11(7): 1795-9, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27128111

RESUMEN

Nurr1/NR4A2 is an orphan nuclear receptor, and currently there are no known natural ligands that bind Nurr1. A recent metabolomics study identified unsaturated fatty acids, including arachidonic acid and docosahexaenoic acid (DHA), that interact with the ligand-binding domain (LBD) of a related orphan receptor, Nur77/NR4A1. However, the binding location and whether these ligands bind other NR4A receptors were not defined. Here, we show that unsaturated fatty acids also interact with the Nurr1 LBD, and solution NMR spectroscopy reveals the binding epitope of DHA at its putative ligand-binding pocket. Biochemical assays reveal that DHA-bound Nurr1 interacts with high affinity with a peptide derived from PIASγ, a protein that interacts with Nurr1 in cellular extracts, and DHA also affects cellular Nurr1 transactivation. This work is the first structural report of a natural ligand binding to a canonical NR4A ligand-binding pocket and indicates a natural ligand can bind and affect Nurr1 function.


Asunto(s)
Ácidos Grasos Insaturados/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Sitios de Unión , Espectroscopía de Resonancia Magnética
18.
Antimicrob Agents Chemother ; 60(4): 2195-208, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26810656

RESUMEN

The human immunodeficiency virus type 1 (HIV-1) capsid plays crucial roles in HIV-1 replication and thus represents an excellent drug target. We developed a high-throughput screening method based on a time-resolved fluorescence resonance energy transfer (HTS-TR-FRET) assay, using the C-terminal domain (CTD) of HIV-1 capsid to identify inhibitors of capsid dimerization. This assay was used to screen a library of pharmacologically active compounds, composed of 1,280in vivo-active drugs, and identified ebselen [2-phenyl-1,2-benzisoselenazol-3(2H)-one], an organoselenium compound, as an inhibitor of HIV-1 capsid CTD dimerization. Nuclear magnetic resonance (NMR) spectroscopic analysis confirmed the direct interaction of ebselen with the HIV-1 capsid CTD and dimer dissociation when ebselen is in 2-fold molar excess. Electrospray ionization mass spectrometry revealed that ebselen covalently binds the HIV-1 capsid CTD, likely via a selenylsulfide linkage with Cys198 and Cys218. This compound presents anti-HIV activity in single and multiple rounds of infection in permissive cell lines as well as in primary peripheral blood mononuclear cells. Ebselen inhibits early viral postentry events of the HIV-1 life cycle by impairing the incoming capsid uncoating process. This compound also blocks infection of other retroviruses, such as Moloney murine leukemia virus and simian immunodeficiency virus, but displays no inhibitory activity against hepatitis C and influenza viruses. This study reports the use of TR-FRET screening to successfully identify a novel capsid inhibitor, ebselen, validating HIV-1 capsid as a promising target for drug development.


Asunto(s)
Fármacos Anti-VIH/farmacología , Azoles/farmacología , Proteínas de la Cápside/antagonistas & inhibidores , Cápside/efectos de los fármacos , VIH-1/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Fármacos Anti-VIH/química , Azoles/química , Sitios de Unión , Cápside/química , Cápside/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Bases de Datos Farmacéuticas , Transferencia Resonante de Energía de Fluorescencia , VIH-1/fisiología , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Isoindoles , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/virología , Virus de la Leucemia Murina de Moloney/efectos de los fármacos , Virus de la Leucemia Murina de Moloney/fisiología , Compuestos de Organoselenio/química , Unión Proteica , Dominios Proteicos , Multimerización de Proteína/efectos de los fármacos , Estructura Secundaria de Proteína , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Virus de la Inmunodeficiencia de los Simios/fisiología , Bibliotecas de Moléculas Pequeñas/química , Ensamble de Virus/efectos de los fármacos , Ensamble de Virus/fisiología , Replicación Viral/efectos de los fármacos
19.
Phys Chem Chem Phys ; 18(8): 5819-31, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26489725

RESUMEN

The conformational landscape of HIV-1 protease (PR) can be experimentally characterized by pulsed-EPR double electron-electron resonance (DEER). For this characterization, nitroxide spin labels are attached to an engineered cysteine residue in the flap region of HIV-1 PR. DEER distance measurements from spin-labels contained within each flap of the homodimer provide a detailed description of the conformational sampling of apo-enzyme as well as induced conformational shifts as a function of inhibitor binding. The distance distribution profiles are further interpreted in terms of a conformational ensemble scheme that consists of four unique states termed "curled/tucked", "closed", "semi-open" and "wide-open" conformations. Reported here are the DEER results for a drug-resistant variant clinical isolate sequence, V6, in the presence of FDA approved protease inhibitors (PIs) as well as a non-hydrolyzable substrate mimic, CaP2. Results are interpreted in the context of the current understanding of the relationship between conformational sampling, drug resistance, and kinetic efficiency of HIV-1PR as derived from previous DEER and kinetic data for a series of HIV-1PR constructs that contain drug-pressure selected mutations or natural polymorphisms. Specifically, these collective results support the notion that inhibitor-induced closure of the flaps correlates with inhibitor efficiency and drug resistance. This body of work also suggests DEER as a tool for studying conformational sampling in flexible enzymes as it relates to function.


Asunto(s)
Espectroscopía de Resonancia por Spin del Electrón , Proteasa del VIH/química , VIH-1/química , Secuencia de Aminoácidos , Clonación Molecular , Resistencia a Medicamentos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteasa del VIH/genética , VIH-1/efectos de los fármacos , Humanos , Modelos Moleculares , Conformación Proteica
20.
Proc Natl Acad Sci U S A ; 113(2): 326-31, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26715749

RESUMEN

Many genomes contain families of paralogs--proteins with divergent function that evolved from a common ancestral gene after a duplication event. To understand how paralogous transcription factors evolve divergent DNA specificities, we examined how the glucocorticoid receptor and its paralogs evolved to bind activating response elements [(+)GREs] and negative glucocorticoid response elements (nGREs). We show that binding to nGREs is a property of the glucocorticoid receptor (GR) DNA-binding domain (DBD) not shared by other members of the steroid receptor family. Using phylogenetic, structural, biochemical, and molecular dynamics techniques, we show that the ancestral DBD from which GR and its paralogs evolved was capable of binding both nGRE and (+)GRE sequences because of the ancestral DBD's ability to assume multiple DNA-bound conformations. Subsequent amino acid substitutions in duplicated daughter genes selectively restricted protein conformational space, causing this dual DNA-binding specificity to be selectively enhanced in the GR lineage and lost in all others. Key substitutions that determined the receptors' response element-binding specificity were far from the proteins' DNA-binding interface and interacted epistatically to change the DBD's function through DNA-induced allosteric mechanisms. These amino acid substitutions subdivided both the conformational and functional space of the ancestral DBD among the present-day receptors, allowing a paralogous family of transcription factors to control disparate transcriptional programs despite high sequence identity.


Asunto(s)
ADN/metabolismo , Homología de Secuencia de Aminoácido , Factores de Transcripción/metabolismo , Regulación Alostérica , Sustitución de Aminoácidos , Secuencia de Bases , Células HeLa , Humanos , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/metabolismo , Elementos de Respuesta/genética , Especificidad por Sustrato , Factores de Transcripción/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...