Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomed Pharmacother ; 152: 113230, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35687908

RESUMEN

BACKGROUND: Bromodomain and extraterminal proteins (BETs) are more than just epigenetic regulators of transcription. Here we highlight a new role for the BET protein BRD4 in the maintenance of higher order chromatin structure at Topologically Associating Domain Boundaries (TADBs). BD2-selective and pan (non-selective) BET inhibitors (BETi) differentially support chromatin structure, selectively affecting transcription and cell viability. METHODS: Using RNA-seq and BRD4 ChIP-seq, the differential effect of BETi treatment on the transcriptome and BRD4 chromatin occupancy of human aortic endothelial cells from diabetic patients (dHAECs) stimulated with TNFα was evaluated. Chromatin decondensation and DNA fragmentation was assessed by immunofluorescence imaging and quantification. Key dHAEC findings were verified in proliferating monocyte-like THP-1 cells using real time-PCR, BRD4 co-immunoprecipitation studies, western blots, proliferation and apoptosis assays. FINDINGS: We discovered that 1) BRD4 co-localizes with Ying-Yang 1 (YY1) at TADBs, critical chromatin structure complexes proximal to many DNA repair genes. 2) BD2-selective BETi enrich BRD4/YY1 associations, while pan-BETi do not. 3) Failure to support chromatin structures through BRD4/YY1 enrichment inhibits DNA repair gene transcription, which induces DNA damage responses, and causes widespread chromatin decondensation, DNA fragmentation, and apoptosis. 4) BD2-selective BETi maintain high order chromatin structure and cell viability, while reducing deleterious pro-inflammatory transcription. INTERPRETATION: BRD4 plays a previously unrecognized role at TADBs. BETi differentially impact TADB stability. Our results provide translational insight for the development of BETi as therapeutics for a range of diseases including CVD, chronic kidney disease, cancer, and COVID-19.


Asunto(s)
COVID-19 , Factores de Transcripción , Proteínas de Ciclo Celular/metabolismo , Cromatina , Células Endoteliales/metabolismo , Epigénesis Genética , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo
2.
Cancer Gene Ther ; 29(6): 859-869, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34385584

RESUMEN

CDK4/6 inhibitors significantly prolong progression-free survival in patients with advanced hormone receptor-positive (HR+) HER2-negative breast cancer. Despite recent successes, patients acquire resistance, necessitating the development of additional novel therapeutic strategies. Bromodomain and extra-terminal domain (BET) proteins are key epigenetic regulators that interact with acetylated lysine (AcLys) residues of histones or transcription factors. BET proteins are directly involved in modulating estrogen receptor (ER) signaling and the cell cycle. Therefore, BET inhibitors can potentially offer new strategies in the treatment of advanced ER+ breast cancer. ZEN-3694 is an orally bioavailable small molecule BET inhibitor currently being evaluated in Phase 1/2 clinical trials (NCT03901469). To assess a potential combination strategy in a CDK4/6i resistant breast cancer population, we investigated the mechanism of action of ZEN-3694 combined with CDK4/6 inhibitors in the ER+ cell lines resistant to palbociclib or abemaciclib. Here, we describe that the combination of ZEN-3694 with CDK4/6i potently inhibits proliferation and induces apoptosis in CDK4/6i resistant cell lines. The resistance to both palbociclib and abemaciclib was associated with the strong upregulation of CDK6 and CCND1 protein levels, which was reversed by the ZEN-3694 treatment. Furthermore, RNAseq data and pathway analysis elucidated the combinatorial effects of ZEN-3694 with CDK4/6 inhibitors through significant downregulation of multiple pathways involved in cell cycle regulation, cellular growth, proliferation, apoptosis, inflammation, and cellular immune response. Our data indicate that ZEN-3694 has therapeutic potential in combination with CDK4/6 inhibitors in patients with advanced ER+ breast resistant to CDK4/6 inhibitors.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/uso terapéutico , Femenino , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
3.
ACS Omega ; 6(33): 21327-21338, 2021 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-34471737

RESUMEN

PB1 is a bromodomain-containing protein hypothesized to act as the nucleosome-recognition subunit of the PBAF complex. Although PB1 is a key component of the PBAF chromatin remodeling complex, its exact role has not been elucidated due to the lack of potent and selective inhibitors. Chemical probes that target specific bromodomains within the complex would constitute highly valuable tools to characterize the function and therapeutic pertinence of PB1 and of each of its bromodomains. Here, we report the design and synthesis of lead compound LM146, which displays strong stabilization of the second and fifth bromodomains of PB1 as shown by DSF. LM146 does not interact with bromodomains outside of sub-family VIII and binds to PB1(2), PB1(5), and SMARCA2B with K D values of 110, 61, and 2100 nM, respectively, providing a ∼34-fold selectivity profile for PB1(5) over SMARCA2.

4.
Bioorg Med Chem Lett ; 47: 128208, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34146702

RESUMEN

BPTF (bromodomain and PHD finger containing transcription factor) is a multidomain protein that plays essential roles in transcriptional regulation, T-cell homeostasis and stem cell pluripotency. As part of the chromatin remodeling complex hNURF (nucleosome remodeling factor), BPTF epigenetic reader subunits are particularly important for BPTF cellular function. Here we report the synthesis of NVS-BPTF-1, a previously reported highly potent and selective BPTF-bromodomain inhibitor. Evaluation of the impact of the inhibition of BPTF-bromodomain using NVS-BPTF-1 on selected proteins involved in the antigen processing pathway revealed that exclusively targeting BPTF-bromodomain is insufficient to observe an increase of PSMB8, PSMB9, TAP1 and TAP2 proteins.


Asunto(s)
Proteínas del Tejido Nervioso/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Antígenos Nucleares , Relación Dosis-Respuesta a Droga , Humanos , Estructura Molecular , Relación Estructura-Actividad
5.
J Med Chem ; 61(18): 8202-8211, 2018 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-30165024

RESUMEN

BET proteins are key epigenetic regulators that regulate transcription through binding to acetylated lysine (AcLys) residues of histones and transcription factors through bromodomains (BDs). The disruption of this interaction with small molecule bromodomain inhibitors is a promising approach to treat various diseases including cancer, autoimmune and cardiovascular diseases. Covalent inhibitors can potentially offer a more durable target inhibition leading to improved in vivo pharmacology. Here we describe the design of covalent inhibitors of BRD4(BD1) that target a methionine in the binding pocket by attaching an epoxide warhead to a suitably oriented noncovalent inhibitor. Using thermal denaturation, MALDI-TOF mass spectrometry, and an X-ray crystal structure, we demonstrate that these inhibitors selectively form a covalent bond with Met149 in BRD4(BD1) but not other bromodomains and provide durable transcriptional and antiproliferative activity in cell based assays. Covalent targeting of methionine offers a novel approach to drug discovery for BET proteins and other targets.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Descubrimiento de Drogas , Neoplasias Hematológicas/tratamiento farmacológico , Metionina/química , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Antineoplásicos/química , Proteínas de Ciclo Celular , Cristalografía por Rayos X , Neoplasias Hematológicas/patología , Humanos , Modelos Moleculares , Estructura Molecular , Conformación Proteica , Relación Estructura-Actividad , Células Tumorales Cultivadas
6.
Drug Discov Today Technol ; 24: 19-24, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29233295

RESUMEN

Inhibition of bromo and extra-terminal (BET) bromodomains, including BRD4, has emerged as a new exciting epigenetic target for oncology, in particular. Recently, novel alternatives to the traditional use of reversible small molecules have emerged, including proteolytic targeting BET agents and irreversible binding inhibitors. These alternatives to reversible inhibitors may offer some advantage and can be used as tools to further decipher the underlying biology. Supportive pre-clinical data have these novel approaches bound for clinical development in the near future.


Asunto(s)
Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular , Descubrimiento de Drogas , Humanos , Proteínas Nucleares/metabolismo , Proteínas/antagonistas & inhibidores , Proteínas/metabolismo , Factores de Transcripción/metabolismo
7.
Biochem Biophys Res Commun ; 477(1): 62-67, 2016 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-27282480

RESUMEN

Bromodomains are epigenetic readers that specifically bind to the acetyl lysine residues of histones and transcription factors. Small molecule BET bromodomain inhibitors can disrupt this interaction which leads to potential modulation of several disease states. Here we describe the binding properties of a novel BET inhibitor RVX-297 that is structurally related to the clinical compound RVX-208, currently undergoing phase III clinical trials for the treatment of cardiovascular diseases, but is distinctly different in its biological and pharmacokinetic profiles. We report that RVX-297 preferentially binds to the BD2 domains of the BET bromodomain and Extra Terminal (BET) family of protein. We demonstrate the differential binding modes of RVX-297 in BD1 and BD2 domains of BRD4 and BRD2 using X-ray crystallography, and describe the structural differences driving the BD2 selective binding of RVX-297. The isothermal titration calorimetry (ITC) data illustrate the related differential thermodynamics of binding of RVX-297 to single as well as dual BET bromodomains.


Asunto(s)
Quinazolinonas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Sitios de Unión , Calorimetría , Cristalografía por Rayos X , Termodinámica , Factores de Transcripción/química
8.
Bioorg Med Chem Lett ; 25(14): 2818-23, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26022843

RESUMEN

Bromodomains are key transcriptional regulators that are thought to be druggable epigenetic targets for cancer, inflammation, diabetes and cardiovascular therapeutics. Of particular importance is the first of two bromodomains in bromodomain containing 4 protein (BRD4(1)). Protein-ligand docking in BRD4(1) was used to purchase a small, focused screening set of compounds possessing a large variety of core structures. Within this set, a small number of weak hits each contained a dihydroquinoxalinone ring system. We purchased other analogs with this ring system and further validated the new hit series and obtained improvement in binding inhibition. Limited exploration by new analog synthesis showed that the binding inhibition in a FRET assay could be improved to the low µM level making this new core a potential hit-to-lead series. Additionally, the predicted geometries of the initial hit and an improved analog were confirmed by X-ray co-crystallography with BRD4(1).


Asunto(s)
Diseño de Fármacos , Ligandos , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Sitios de Unión , Proteínas de Ciclo Celular , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Humanos , Simulación del Acoplamiento Molecular , Proteínas Nucleares/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Quinoxalinas/química , Quinoxalinas/metabolismo , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
9.
J Biochem ; 154(4): 383-91, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23975754

RESUMEN

Abscisic acid (ABA) is a stress-inducible plant hormone comprising an inevitable component of the human diet. Recently, stress-induced accumulation of autocrine ABA was shown in humans, as well as ABA-mediated modulation of a number of disease-associated systems. Now, the application of a chemical proteomics approach to gain further insight into ABA mechanisms of action in mammalian cells is reported. An ABA mimetic photoaffinity probe was applied to intact mammalian insulinoma and embryonic cells, leading to the identification of heat shock protein 70 (HSP70) family members, (including GRP78 and HSP70-2) as putative human ABA-binding proteins. In vitro characterization of the ABA-HSP70 interactions yielded K(d)s in the 20-60 µM range, which decreased several fold in the presence of co-chaperone. However, ABA was found to have only variable- and co-chaperone-independent effects on the ATPase activity of these proteins. The potential implications of these ABA-HSP70 interactions are discussed with respect to the intracellular protein folding and extracellular receptor-like activities of these stress-inducible proteins. While mechanistic and functional relevance remain enigmatic, we conclude that ABA can bind to human HSP70 family members with physiologically relevant affinities and in a co-chaperone-dependent manner.


Asunto(s)
Ácido Abscísico/metabolismo , Proteínas del Choque Térmico HSP40/metabolismo , Proteínas de Choque Térmico/metabolismo , Animales , Línea Celular Tumoral , Chaperón BiP del Retículo Endoplásmico , Células HEK293 , Humanos , Unión Proteica , Proteómica , Ratas
10.
PLoS Comput Biol ; 9(6): e1003114, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825939

RESUMEN

The pyrabactin resistance 1 (PYR1)/PYR1-like (PYL)/regulatory component of abscisic acid (ABA) response (RCAR) proteins comprise a well characterized family of ABA receptors. Recent investigations have revealed two subsets of these receptors that, in the absence of ABA, either form inactive homodimers (PYR1 and PYLs 1-3) or mediate basal inhibition of downstream target type 2C protein phosphatases (PP2Cs; PYLs 4-10) respectively in vitro. Addition of ABA has been shown to release the apo-homodimers yielding ABA-bound monomeric holo-receptors that can interact with PP2Cs; highlighting a competitive-interaction process. Interaction selectivity has been shown to be mediated by subtle structural variations of primary sequence and ligand binding effects. Now, the dynamical contributions of ligand binding on interaction selectivity are investigated through extensive molecular dynamics (MD) simulations of apo and holo-PYR1 in monomeric and dimeric form as well as in complex with a PP2C, homology to ABA insensitive 1 (HAB1). Robust comparative interpretations were enabled by a novel essential collective dynamics approach. In agreement with recent experimental findings, our analysis indicates that ABA-bound PYR1 should efficiently bind to HAB1. However, both ABA-bound and ABA-extracted PYR1-HAB1 constructs have demonstrated notable similarities in their dynamics, suggesting that apo-PYR1 should also be able to make a substantial interaction with PP2Cs, albeit likely with slower complex formation kinetics. Further analysis indicates that both ABA-bound and ABA-free PYR1 in complex with HAB1 exhibit a higher intra-molecular structural stability and stronger inter-molecular dynamic correlations, in comparison with either holo- or apo-PYR1 dimers, supporting a model that includes apo-PYR1 in complex with HAB1. This possibility of a conditional functional apo-PYR1-PP2C complex was validated in vitro. These findings are generally consistent with the competitive-interaction model for PYR1 but highlight dynamical contributions of the PYR1 structure in mediating interaction selectivity suggesting added degrees of complexity in the regulation of the competitive-inhibition.


Asunto(s)
Ácido Abscísico/metabolismo , Proteínas de Arabidopsis/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Arabidopsis/metabolismo , Cinética , Ligandos , Simulación de Dinámica Molecular , Unión Proteica
11.
Plant Physiol Biochem ; 68: 32-6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23624020

RESUMEN

The G-protein coupled receptor-type G-proteins (GTG) 1 and 2 from Arabidopsis thaliana have been proposed to function in the modulation of abscisic acid (ABA) mediated responses to stress and development. In particular it has been suggested that they function as ABA receptors based on in planta and in vitro analyses. However a recent independent report was inconsistent with this, suggesting that there is no link between the GTGs and ABA in planta. Here we provide an independent assessment of the ability of ABA to bind to recombinant GTG1 in vitro and in vivo in Sacaromycese cerevisiae. Radio-labelled binding assays on enriched lipid-reconstituted recombinant GTG1, demonstrated specific concentration dependent binding of [(3)H]-ABA with a dissociation constant (KD) of 80 nM, corroborating previous reports. Assessment of the binding of [(3)H]-ABA to intact GTG1 expressing yeast, showed GTG1-dependent binding in vivo, yielding a physiologically relevant KD of 0.6 µM. Together these results provide independent evidence of a binding-interaction between ABA and GTG1 in vitro and in vivo, in support of the previously proposed possibility of a biologically relevant interaction between GTG1 and ABA.


Asunto(s)
Ácido Abscísico/metabolismo , Proteínas de Arabidopsis/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Arabidopsis/genética , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/metabolismo
12.
PLoS One ; 8(12): e83190, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24391744

RESUMEN

Increased synthesis of Apolipoprotein A-I (ApoA-I) and HDL is believed to provide a new approach to treating atherosclerosis through the stimulation of reverse cholesterol transport. RVX-208 increases the production of ApoA-I in hepatocytes in vitro, and in vivo in monkeys and humans, which results in increased HDL-C, but the molecular target was not previously reported. Using binding assays and X-ray crystallography, we now show that RVX-208 selectively binds to bromodomains of the BET (Bromodomain and Extra Terminal) family, competing for a site bound by the endogenous ligand, acetylated lysine, and that this accounts for its pharmacological activity. siRNA experiments further suggest that induction of ApoA-I mRNA is mediated by BET family member BRD4. These data indicate that RVX-208 increases ApoA-I production through an epigenetic mechanism and suggests that BET inhibition may be a promising new approach to the treatment of atherosclerosis.


Asunto(s)
Apolipoproteína A-I/biosíntesis , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Quinazolinas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Animales , Apolipoproteína A-I/genética , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Sitios de Unión , Proteínas de Ciclo Celular , Línea Celular , Cristalografía por Rayos X , Epigénesis Genética/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/genética , Conformación Proteica , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Quinazolinas/química , Quinazolinonas , ARN Interferente Pequeño/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Factores de Transcripción/química , Factores de Transcripción/genética
13.
Biochem J ; 437(1): 117-23, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21473740

RESUMEN

ABA (abscisic acid) is a plant hormone involved in important processes including development and stress responses. Recent reports have identified a number of plant ABA receptors and transporters, highlighting novel mechanisms of ABA action. In the present paper we describe application of a chemical proteomics approach leading to the identification of mitochondrial ANTs (adenine nucleotide translocators) as ABA-interacting proteins. Initial in vitro studies confirmed inhibition of ANT-dependent ATP translocation by ABA. Further analysis demonstrated ANT-dependent uptake of ABA into both recombinant Arabidopsis thaliana ANT2-containing proteoliposomes and native isolated spinach mitochondria; the latter with a Km of 3.5 µM and a Vmax of 2.5 nmol/min per g of protein. ATP was found to inhibit ANT-dependent ABA translocation. Specificity profiles highlight the possibility of mechanistic differences in translocation of ABA and ATP. Finally, ABA was shown to stimulate ATPase activity in spinach mitochondrial extracts. ABA concentrations in plant cells are estimated to reach the low micromolar range during stress responses, supporting potential physiological relevance of these in vitro findings. Overall, the present in vitro work suggests the possibility of as yet uncharacterized mechanisms of ABA action in planta related to inhibition of mitochondrial ATP translocation and functional localization of ABA in the mitochondrial matrix.


Asunto(s)
Ácido Abscísico/metabolismo , Translocador 2 del Nucleótido Adenina/metabolismo , Nucleótidos de Adenina/metabolismo , Mitocondrias/metabolismo , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/metabolismo , Arabidopsis/metabolismo , Proteolípidos/metabolismo , Transducción de Señal
14.
Inorg Chem ; 45(25): 9974-84, 2006 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-17140193

RESUMEN

The alpha-helical coiled-coil motif serves as a robust scaffold for incorporating electron-transfer (ET) functionality into synthetic metalloproteins. These structures consist of a supercoiling of two or more aplha helices that are formed by the self-assembly of individual polypeptide chains whose sequences contain a repeating pattern of hydrophobic and hydrophilic residues. Early work from our group attached abiotic Ru-based redox sites to the most surface-exposed positions of two stranded coiled-coils and used electron-pulse radiolysis to study both intra- and intermolecular ET reactions in these systems. Later work used smaller metallopeptides to investigate the effects of conformational gating within electrostatic peptide-protein complexes. We have recently designed the C16C19-GGY peptide, which contains Cys residues located at both the "a" and "d" positions of its third heptad repeat in order to construct a nativelike metal-binding domain within its hydrophobic core. It was shown that the binding of both Cd(II) and Cu(I) ions induces the peptide to undergo a conformational change from a disordered random coil to a metal-bridged coiled-coil. However, whereas the Cd(II)-protein exists as a two-stranded coiled-coil, the Cu(I) derivative exists as a four-stranded coiled-coil. Upon the incorporation of other metal ions, metal-bridged peptide dimers, tetramers, and hexamers are formed. The Cu(I)-protein is of particular interest because it exhibits a long-lived (microsecond) room-temperature luminescence at 600 nm. The luminophore in this protein is thought to be a multinuclear CuI4Cys4(N/O)4 cage complex, which can be quenched by exogenous electron acceptors in solution, as shown by emission-lifetime and transient-absorption experiments. It is anticipated that further investigation into these systems will contribute to the expanding effort of bioinorganic chemists to prepare new kinds of functionally active synthetic metalloproteins.


Asunto(s)
Transporte de Electrón/fisiología , Metaloproteínas/química , Metales/química , Péptidos/química , Secuencia de Aminoácidos , Sitios de Unión , Cadmio/química , Simulación por Computador , Cobre/química , Cisteína/química , Cisteína/metabolismo , Dimerización , Interacciones Hidrofóbicas e Hidrofílicas , Metaloproteínas/metabolismo , Datos de Secuencia Molecular , Oxidación-Reducción , Péptidos/metabolismo , Estructura Secundaria de Proteína , Rutenio/química , Electricidad Estática , Factores de Tiempo , Viscosidad
16.
J Am Chem Soc ; 127(21): 7678-9, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15913348

RESUMEN

The addition of Cu(I) to the random-coil peptide, C16C19-GGY, produces a self-organized, metal-bridged 4-helix bundle which displays an intense room-temperature luminescence at 600 nm. Emission, UV, and CD titrations along with X-ray absorption studies indicate that the luminescent cofactor is likely a Cu4S4 cluster in which each Cu atom is bridged by the side chains of two cysteine residues and has terminal N/O ligation.


Asunto(s)
Cobre/química , Metaloproteínas/química , Sulfuros/química , Secuencia de Aminoácidos , Dicroismo Circular , Análisis de Fourier , Mediciones Luminiscentes , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Análisis Espectral
17.
J Inorg Biochem ; 98(11): 1971-4, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15522423

RESUMEN

The metal-induced assembly of a designed peptide-based rubredoxin model is described. The C16C19-GGY peptide has the sequence Ac-K(IEALEGK)(2)(CEACEGK)(IEALEGK)GGY-amide in which the presence of the Cys-X-X-Cys metal binding domain of rubredoxin was used to place cysteine residues at the hydrophobic "a" and "d" positions upon formation of a homodimeric alpha-helical coiled-coil. Circular dichroism spectroscopy shows that the apopeptide exists as a random coil and assembles into a coiled-coil in the presence of Cd(2+). Metal binding is monitored by the appearance of a new LMCT band at 238 nm. UV-Vis titrations and SDS-PAGE experiments are used to show that this designed metalloprotein exists as a metal-bridged coiled-coil dimer.


Asunto(s)
Cisteína , Metaloproteínas/química , Metales/química , Secuencia de Aminoácidos , Cadmio , Simulación por Computador , Dimerización , Metaloproteínas/efectos de los fármacos , Metaloproteínas/metabolismo , Metales/farmacología , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos/química , Conformación Proteica , Pliegue de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...