Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Med Chem ; 64(24): 18193-18208, 2021 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-34894681

RESUMEN

As a result of emerging biological data suggesting that within the c-Jun N-terminal kinase (JNK) family, JNK1 and not JNK2 or JNK3 may be primarily responsible for fibrosis pathology, we sought to identify JNK inhibitors with an increased JNK1 bias relative to our previous clinical compound tanzisertib (CC-930). This manuscript reports the synthesis and structure-activity relationship (SAR) studies for a novel series of JNK inhibitors demonstrating an increased JNK1 bias. SAR optimization on a series of 2,4-dialkylamino-pyrimidine-5-carboxamides resulted in the identification of compounds possessing low nanomolar JNK inhibitory potency, overall kinome selectivity, and the ability to inhibit cellular phosphorylation of the direct JNK substrate c-Jun. Optimization of physicochemical properties in this series resulted in compounds that demonstrated excellent systemic exposure following oral dosing, enabling in vivo efficacy studies and the selection of a candidate for clinical development, CC-90001, which is currently in clinical trials (Phase II) in patients with idiopathic pulmonary fibrosis (NCT03142191).


Asunto(s)
Ciclohexilaminas/farmacología , Descubrimiento de Drogas , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Animales , Ciclohexilaminas/uso terapéutico , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Fosforilación , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas/uso terapéutico , Relación Estructura-Actividad , Especificidad por Sustrato
2.
J Med Chem ; 64(17): 12670-12679, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34459599

RESUMEN

TTK is an essential spindle assembly checkpoint enzyme in many organisms. It plays a central role in tumor cell proliferation and is aberrantly overexpressed in a wide range of tumor types. We recently reported on a series of potent and selective TTK inhibitors with strong antiproliferative activity in triple negative breast cancer (TNBC) cell lines (8: TTK IC50 = 3.0 nM; CAL-51 IC50 = 84.0 nM). Inspired by previously described potent tricyclic TTK inhibitor 6 (TTK IC50 = 0.9 nM), we embarked on a structure-enabled design and optimization campaign to identify an improved series with excellent potency, TTK selectivity, solubility, CYP inhibition profile, and in vivo efficacy in a TNBC xenograft model. These efforts culminated in the discovery of 25 (TTK IC50 = 3.0 nM; CAL-51 IC50 = 16.0 nM), which showed significant single-agent efficacy when dosed iv in a TNBC xenograft model without body weight loss.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Antineoplásicos , Neoplasias de la Mama , Línea Celular Tumoral , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Relación Estructura-Actividad
3.
Drug Metab Lett ; 10(2): 144-50, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27165340

RESUMEN

BACKGROUND: The study of novel sites of metabolism is important in understanding new mechanisms of biotransformation of a particular moiety by metabolic enzymes. This information is valuable in designing metabolically-stable compounds with drug-like properties. It may also provide insights into the existence of active and reactive metabolites. METHODS: We utilized small scale incubations to generate adequate amounts of the metabolite of interest. After purification, LC-MS/MS and Proton Nuclear Magnetic Resonance (1H-NMR) were utilized to unequivocally assign the novel site of glutathione conjugation on the purine ring system. RESULTS: A proposed novel site of glutathione conjugation was investigated on a diaminopurine-containing molecule. It was demonstrated that the formation of the glutathione conjugate at the C-6 position of the purine ring system was due to the bioactivation of the compound to a di-imine intermediate by CYP3A4, followed by the nucleophilic addition of glutathione. CONCLUSION: S-glutathionylation at C-6 position of a purine was proven unequivocally. This previously unreported mechanism constitutes a novel biotransformation for purines.


Asunto(s)
Cromatografía Liquida/métodos , Citocromo P-450 CYP3A/metabolismo , Glutatión/metabolismo , Purinas/metabolismo , Animales , Perros , Haplorrinos , Humanos , Espectroscopía de Resonancia Magnética/métodos , Ratones , Ratas , Espectrometría de Masas en Tándem
4.
J Med Chem ; 58(13): 5323-33, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-26083478

RESUMEN

We report here the synthesis and structure-activity relationship (SAR) of a novel series of mammalian target of rapamycin (mTOR) kinase inhibitors. A series of 4,6- or 1,7-disubstituted-3,4-dihydropyrazino[2,3-b]pyrazine-2(1H)-ones were optimized for in vivo efficacy. These efforts resulted in the identification of compounds with excellent mTOR kinase inhibitory potency, with exquisite kinase selectivity over the related lipid kinase PI3K. The improved PK properties of this series allowed for exploration of in vivo efficacy and ultimately the selection of CC-223 for clinical development.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas , Inhibidores de las Quinasa Fosfoinosítidos-3 , Neoplasias de la Próstata/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Antineoplásicos/síntesis química , Humanos , Masculino , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Pirazinas/síntesis química , Ratas , Relación Estructura-Actividad , Células Tumorales Cultivadas
5.
J Med Chem ; 58(14): 5599-608, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26102506

RESUMEN

We report here the synthesis and structure-activity relationship (SAR) of a novel series of triazole containing mammalian target of rapamycin (mTOR) kinase inhibitors. SAR studies examining the potency, selectivity, and PK parameters for a series of triazole containing 4,6- or 1,7-disubstituted-3,4-dihydropyrazino[2,3-b]pyrazine-2(1H)-ones resulted in the identification of triazole containing mTOR kinase inhibitors with improved PK properties. Potent compounds from this series were found to block both mTORC1(pS6) and mTORC2(pAktS473) signaling in PC-3 cancer cells, in vitro and in vivo. When assessed in efficacy models, analogs exhibited dose-dependent efficacy in tumor xenograft models. This work resulted in the selection of CC-115 for clinical development.


Asunto(s)
Diseño de Fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/química , Pirazinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Triazoles/química , Triazoles/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Simulación del Acoplamiento Molecular , Conformación Proteica , Inhibidores de Proteínas Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacocinética , Pirazinas/metabolismo , Pirazinas/farmacocinética , Ratas , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Serina-Treonina Quinasas TOR/química , Serina-Treonina Quinasas TOR/metabolismo , Triazoles/metabolismo , Triazoles/farmacocinética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Cancer Ther ; 14(6): 1295-305, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25855786

RESUMEN

mTOR is a serine/threonine kinase that regulates cell growth, metabolism, proliferation, and survival. mTOR complex-1 (mTORC1) and mTOR complex-2 (mTORC2) are critical mediators of the PI3K-AKT pathway, which is frequently mutated in many cancers, leading to hyperactivation of mTOR signaling. Although rapamycin analogues, allosteric inhibitors that target only the mTORC1 complex, have shown some clinical activity, it is hypothesized that mTOR kinase inhibitors, blocking both mTORC1 and mTORC2 signaling, will have expanded therapeutic potential. Here, we describe the preclinical characterization of CC-223. CC-223 is a potent, selective, and orally bioavailable inhibitor of mTOR kinase, demonstrating inhibition of mTORC1 (pS6RP and p4EBP1) and mTORC2 [pAKT(S473)] in cellular systems. Growth inhibitory activity was demonstrated in hematologic and solid tumor cell lines. mTOR kinase inhibition in cells, by CC-223, resulted in more complete inhibition of the mTOR pathway biomarkers and improved antiproliferative activity as compared with rapamycin. Growth inhibitory activity and apoptosis was demonstrated in a panel of hematologic cancer cell lines. Correlative analysis revealed that IRF4 expression level associates with resistance, whereas mTOR pathway activation seems to associate with sensitivity. Treatment with CC-223 afforded in vivo tumor biomarker inhibition in tumor-bearing mice, after a single oral dose. CC-223 exhibited dose-dependent tumor growth inhibition in multiple solid tumor xenografts. Significant inhibition of mTOR pathway markers pS6RP and pAKT in CC-223-treated tumors suggests that the observed antitumor activity of CC-223 was mediated through inhibition of both mTORC1 and mTORC2. CC-223 is currently in phase I clinical trials.


Asunto(s)
Neoplasias/tratamiento farmacológico , Pirazinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Células HCT116 , Células HEK293 , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones SCID , Estructura Molecular , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neovascularización Patológica/prevención & control , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirazinas/química , Serina-Treonina Quinasas TOR/metabolismo , Carga Tumoral/efectos de los fármacos
7.
Drug Metab Lett ; 8(1): 19-30, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24628405

RESUMEN

The in vitro and in vivo preclinical ADME properties of 10 clinically late stage or marketed covalent inhibitors were evaluated in order to define advancement criteria for discovery of future drugs in this arena. Our studies revealed the following: After incubating with S9 fractions for 30 minutes, the rat and human in vitro stability for these compounds ranged from 1% to 100%. The blood stability ranged from 30% to 100%. There was a broad range of CYP inhibition with prevalence for time-dependent inhibition of at least one enzyme. The Caco-2 permeability (A→B) ranged from negligible (0.6 x 10(-6) cm/s) to highly permeable (31 x 10(-6) cm/s) and the efflux ratio also varied widely (0.2-30). Most of the compounds were highly protein bound in both rat and human with binding ≥ 90%. Rat plasma clearance for the 10 compounds ranged from slow (11 mL/min/kg) to very rapid (350 mL/min/kg). The Vss ranged from low (0.67 L/kg) to very high (115 L/kg). MRT's also ranged from short (0.5 hr) to long (7.4 hr). The oral exposures also showed a very broad range with CMax's ranging from 0.01-77 µM and exposure levels ranging from 0.03-106 µM.hr. In conclusion, the wide range in in vitro and in vivo ADME data makes these particular ADME assays non-discriminatory in the selection of promising compounds. In our opinion, non-traditional assays such as target mass modification, target confirmation by amino acid sequencing, cellular target occupancy, and target turnover rate data in combination with the pharmacokinetic profiles are the critical considerations for progression of irreversible compounds in early discovery.


Asunto(s)
Inhibidores Enzimáticos del Citocromo P-450/farmacología , Sistema Enzimático del Citocromo P-450/metabolismo , Descubrimiento de Drogas/métodos , Ensayos Analíticos de Alto Rendimiento , Microsomas Hepáticos/efectos de los fármacos , Administración Intravenosa , Administración Oral , Animales , Células CACO-2 , Inhibidores Enzimáticos del Citocromo P-450/administración & dosificación , Inhibidores Enzimáticos del Citocromo P-450/sangre , Inhibidores Enzimáticos del Citocromo P-450/farmacocinética , Estabilidad de Medicamentos , Humanos , Absorción Intestinal , Mucosa Intestinal/metabolismo , Isoenzimas , Cinética , Masculino , Microsomas Hepáticos/enzimología , Modelos Biológicos , Permeabilidad , Unión Proteica , Ratas , Especificidad por Sustrato , Flujo de Trabajo
8.
J Med Chem ; 55(7): 3250-60, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22380736

RESUMEN

Serine/threonine protein kinases Aurora A, B, and C play essential roles in cell mitosis and cytokinesis. Currently a number of Aurora kinase inhibitors with different isoform selectivities are being evaluated in the clinic. Herein we report the discovery and characterization of 21c (AC014) and 21i (AC081), two structurally novel, potent, kinome-selective pan-Aurora inhibitors. In the human colon cancer cell line HCT-116, both compounds potently inhibit histone H3 phosphorylation and cell proliferation while inducing 8N polyploidy. Both compounds administered intravenously on intermittent schedules displayed potent and durable antitumor activity in a nude rat HCT-116 tumor xenograft model and exhibited good in vivo tolerability. Taken together, these data support further development of both 21c and 21i as potential therapeutic agents for the treatment of solid tumors and hematological malignancies.


Asunto(s)
Acetanilidas/síntesis química , Antineoplásicos/síntesis química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Triazinas/síntesis química , Acetanilidas/farmacocinética , Acetanilidas/farmacología , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Aurora Quinasa A , Aurora Quinasas , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Histonas/metabolismo , Humanos , Modelos Moleculares , Trasplante de Neoplasias , Fosforilación , Unión Proteica , Ratas , Ratas Desnudas , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad , Trasplante Heterólogo , Triazinas/farmacocinética , Triazinas/farmacología
9.
Mol Cancer Ther ; 11(4): 930-41, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22319199

RESUMEN

Mutations in the BRAF gene have been identified in approximately 7% of cancers, including 60% to 70% of melanomas, 29% to 83% of papillary thyroid carcinomas, 4% to 16% colorectal cancers, and a lesser extent in serous ovarian and non-small cell lung cancers. The V600E mutation is found in the vast majority of cases and is an activating mutation, conferring transforming and immortalization potential to cells. CEP-32496 is a potent BRAF inhibitor in an in vitro binding assay for mutated BRAF(V600E) (K(d) BRAF(V600E) = 14 nmol/L) and in a mitogen-activated protein (MAP)/extracellular signal-regulated (ER) kinase (MEK) phosphorylation (pMEK) inhibition assay in human melanoma (A375) and colorectal cancer (Colo-205) cell lines (IC(50) = 78 and 60 nmol/L). In vitro, CEP-32496 has multikinase binding activity at other cancer targets of interest; however, it exhibits selective cellular cytotoxicity for BRAF(V600E) versus wild-type cells. CEP-32496 is orally bioavailable in multiple preclinical species (>95% in rats, dogs, and monkeys) and has single oral dose pharmacodynamic inhibition (10-55 mg/kg) of both pMEK and pERK in BRAF(V600E) colon carcinoma xenografts in nude mice. Sustained tumor stasis and regressions are observed with oral administration (30-100 mg/kg twice daily) against BRAF(V600E) melanoma and colon carcinoma xenografts, with no adverse effects. Little or no epithelial hyperplasia was observed in rodents and primates with prolonged oral administration and sustained exposure. CEP-32496 benchmarks favorably with respect to other kinase inhibitors, including RAF-265 (phase I), sorafenib, (approved), and vemurafenib (PLX4032/RG7204, approved). CEP-32496 represents a novel and pharmacologically active BRAF inhibitor with a favorable side effect profile currently in clinical development.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Fenilurea/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Quinazolinas/farmacología , Administración Oral , Animales , Línea Celular Tumoral , Proliferación Celular , Perros , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Desnudos , Proteínas Proto-Oncogénicas B-raf/genética , Quinazolinas/farmacocinética , Ratas , Ratas Sprague-Dawley
10.
Drug Metab Lett ; 6(4): 265-74, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23607719

RESUMEN

Metabolite identification can provide tremendous value in identifying metabolic soft-spots on molecules of interest and to evaluate the potential for generating reactive species. This information is useful in designing stable analogs with acceptable drug-like properties. Two key compounds were found to generate major metabolites that could not be elucidated by mass spectrometry. Nuclear Magnetic Resonance (NMR) is a non-destructive method to obtain structural information. It requires milligram quantities of putative metabolites, typically unavailable in early stage discovery projects. Herein, we demonstrated the application of NMR using microgram quantities of samples to identify the structures of the major metabolites of two discovery compounds. In the first case, we studied structural elucidation of a Nglucuronide on a pyrazole moiety using 1H-NMR due to the instability of the glucuronidated metabolite under mass spectrometric conditions. In the second example, we characterized two oxidized metabolites having identical mass fragmentation using 2D-NMR. In both cases, chemists incorporated these findings into designing analogs to improve metabolic stability.


Asunto(s)
Glucurónidos/química , Espectroscopía de Resonancia Magnética/métodos , Microsomas Hepáticos/metabolismo , Animales , Oxidación-Reducción , Ratas
11.
J Med Chem ; 55(3): 1082-105, 2012 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-22168626

RESUMEN

The Ras/RAF/MEK/ERK mitogen-activated protein kinase (MAPK) signaling pathway plays a central role in the regulation of cell growth, differentiation, and survival. Expression of mutant BRAF(V600E) results in constitutive activation of the MAPK pathway, which can lead to uncontrolled cellular growth. Herein, we describe an SAR optimization campaign around a series of quinazoline derived BRAF(V600E) inhibitors. In particular, the bioisosteric replacement of a metabolically sensitive tert-butyl group with fluorinated alkyl moieties is described. This effort led directly to the identification of a clinical candidate, compound 40 (CEP-32496). Compound 40 exhibits high potency against several BRAF(V600E)-dependent cell lines and selective cytotoxicity for tumor cell lines expressing mutant BRAF(V600E) versus those containing wild-type BRAF. Compound 40 also exhibits an excellent PK profile across multiple preclinical species. In addition, significant oral efficacy was observed in a 14-day BRAF(V600E)-dependent human Colo-205 tumor xenograft mouse model, upon dosing at 30 and 100 mg/kg BID.


Asunto(s)
Isoxazoles/síntesis química , Compuestos de Fenilurea/síntesis química , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Quinazolinas/síntesis química , Administración Oral , Animales , Unión Competitiva , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Perros , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Isoxazoles/farmacocinética , Isoxazoles/farmacología , Macaca fascicularis , Masculino , Ratones , Ratones Desnudos , Microsomas Hepáticos , Modelos Moleculares , Mutación , Trasplante de Neoplasias , Compuestos de Fenilurea/farmacocinética , Compuestos de Fenilurea/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Quinazolinas/farmacocinética , Quinazolinas/farmacología , Ratas , Ratas Sprague-Dawley , Estereoisomerismo , Relación Estructura-Actividad , Trasplante Heterólogo
12.
Biochemistry ; 43(13): 3824-34, 2004 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-15049689

RESUMEN

The coordination structure and reactivities of metal ligands in metal-sensing metalloregulatory coordination complexes may well dictate their biological properties. Here, we use the technique of ratiometric pulsed alkylation mass spectrometry (rPA-MS) to probe the structure and reactivities of metal coordination complexes formed by different metalloderivatives of Staphylococcus aureus plasmid pI258-encoded CadC, the metal-regulated transcriptional repressor of the cad operon. The cad operon provides resistance to large thiophilic heavy metal pollutants including Cd(II), Pb(II), and Bi(III). Two cysteines, an invariant Cys7 and a conserved Cys11, separated by three amino acids near the N-terminus of each subunit within dimeric CadC, donate two of the four coordination bonds to Cd(II) and Bi(III); in contrast, Cys11, but not Cys7, is excluded from the trigonal Pb(II) complex. rPA-MS reveals that Cys7 is strongly protected from alkylation in all metal complexes, Pb(II) being most effective, reducing k(app)(C7)by approximately 1000-fold relative to apo-CadC; in contrast, the reactivity of Cys11 is indistinguishable from that of apo-CadC, consistent with an S(3) coordination complex. Only in the tetrathiolate complexes formed by Cd(II) and Bi(III) is the reactivity of Cys11 appreciably reduced, but only by >or=10-fold. These data suggest that the Cys11-S(-)-metal coordination bond or that side of the coordination chelate in the trigonal Pb(II) complex defines a "weak point" in the chelate and thus might provide an entry site for potential metal ligand exchange reactions important for metal resistance in vivo. In contrast, Cys7 forms a tight coordination bond with all inducing metals, consistent with its role as a critical allosteric ligand in the metalloregulation of the operator/promoter binding.


Asunto(s)
Proteínas Bacterianas/química , Cisteína/química , Metaloproteínas/química , Plásmidos/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Staphylococcus aureus/química , Compuestos de Sulfhidrilo/química , Alquilación , Apoproteínas/química , Apoproteínas/genética , Proteínas Bacterianas/genética , Bismuto/química , Cadmio/química , Quimotripsina/química , Cobalto/química , Etilmaleimida/química , Hidrólisis , Metaloproteínas/genética , Fragmentos de Péptidos/análisis , Fragmentos de Péptidos/química , Staphylococcus aureus/genética , Tripsina/química , Zinc/química
13.
J Biol Inorg Chem ; 7(4-5): 551-9, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11941514

RESUMEN

Staphylococcus aureus pI258 CadC is a metal sensor protein that regulates the expression of the cad operon which encodes metal ion resistance proteins involved in the efficient efflux of Cd(II), Pb(II), Zn(II) and, according to one report, Bi(III) ions. In this paper, direct evidence is presented that Bi(III) binds to CadC and negatively regulates cad operator/promoter (O/P) binding. Optical absorption spectroscopy reveals that dimeric CadC binds approximately 0.8 mol equivalents of Bi(III) per CadC monomer to form a coordination complex characterized by three S(-)-->Bi(III) ligand-to-metal charge transfer transitions, with the longest wavelength absorption band centered at 415 nm (epsilon(415)=4000 M(Bi)(-1) cm(-1)). UV-Vis absorption spectra of wild-type and mutant Cys-->Gly (Ser) substitution CadC mutants compared to [Bi(DTT)(2)], [Bi(GSH)(3)] and [Bi(NAC)](3) model complexes reveal that Cys7, Cys11, Cys60 and Cys58 directly coordinate Bi(III) in a tetrathiolate coordination complex. The apparent affinity derived from a Bi(III)-displacement optical titration with Cd(II) is estimated to be K(Bi)< or =10(12) M(-1). Apo-CadC binds with high affinity [ K(a)=1.1(+/-0.3)x10(9) M(-1); 0.40 M NaCl, pH 7.0, 25 degrees C] to a 5'-fluorescein-labeled cad O/P oligonucleotide,while the binding of one molar equivalent of Bi(III) per CadC monomer (Bi(1)-CadC) reduces the affinity by approximately 170-fold. Strikingly, Bi(III)-responsive negative regulation of cad O/P binding is abrogated for Bi(1)-C60G CadC and severely disrupted in Bi(1)-C7G CadC, whose relative affinity is reduced only 10-fold. The mechanism of Bi(III)-responsive metalloregulation is discussed, based on the findings presented here. Electronic supplementary material to this paper can be obtained by using the Springer Link server located at http://dx.doi.org/10.1007/s00775-001-0336-9.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Bismuto/metabolismo , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Staphylococcus aureus/química , Proteínas Bacterianas/genética , Sitios de Unión , Bismuto/química , Cisteína/genética , Dimerización , Fluorescencia , Regulación Bacteriana de la Expresión Génica , Glicina/genética , Modelos Moleculares , Operón , Mutación Puntual , Regiones Promotoras Genéticas , Conformación Proteica , Espectrofotometría/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA