Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Circ Res ; 111(1): 50-5, 2012 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-22581928

RESUMEN

RATIONALE: Direct reprogramming of fibroblasts into cardiomyocytes is a novel strategy for cardiac regeneration. However, the key determinants involved in this process are unknown. OBJECTIVE: To assess the efficiency of direct fibroblast reprogramming via viral overexpression of GATA4, Mef2c, and Tbx5 (GMT). METHODS AND RESULTS: We induced GMT overexpression in murine tail tip fibroblasts (TTFs) and cardiac fibroblasts (CFs) from multiple lines of transgenic mice carrying different cardiomyocyte lineage reporters. We found that the induction of GMT overexpression in TTFs and CFs is inefficient at inducing molecular and electrophysiological phenotypes of mature cardiomyocytes. In addition, transplantation of GMT infected CFs into injured mouse hearts resulted in decreased cell survival with minimal induction of cardiomyocyte genes. CONCLUSIONS: Significant challenges remain in our ability to convert fibroblasts into cardiomyocyte-like cells and a greater understanding of cardiovascular epigenetics is needed to increase the translational potential of this strategy.


Asunto(s)
Transdiferenciación Celular , Fibroblastos/metabolismo , Miocitos Cardíacos/metabolismo , Factores Reguladores Miogénicos/metabolismo , Proteínas de Dominio T Box/metabolismo , Potenciales de Acción , Animales , Linaje de la Célula , Supervivencia Celular , Transdiferenciación Celular/efectos de los fármacos , Femenino , Fibroblastos/trasplante , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Genotipo , Células HEK293 , Humanos , Factores de Transcripción MEF2 , Masculino , Ratones , Ratones SCID , Ratones Transgénicos , Miocitos Cardíacos/trasplante , Factores Reguladores Miogénicos/genética , Técnicas de Placa-Clamp , Fenotipo , Reacción en Cadena de la Polimerasa , Proteínas de Dominio T Box/genética , Factores de Tiempo , Transcripción Genética , Transfección , Regulación hacia Arriba
2.
Crit Rev Biochem Mol Biol ; 46(4): 295-309, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21539479

RESUMEN

The inhibition of protein kinases has gained general acceptance as an effective approach to treat a wide range of cancers. However, in many cases, prolonged administration of kinase inhibitors often leads to acquired resistance, and the therapeutic effect is subsequently diminished. The wealth of recent studies using biochemical, kinetic, and structural approaches have revealed the molecular basis for the clinically observed resistance. In this review, we highlight several of the most common molecular mechanisms that lead to acquired resistance to kinase inhibitors observed with the cAbl (cellular form of the Abelson leukemia virus tyrosine kinase) and the type III receptor tyrosine kinase cKit, including a newly identified mechanism resulting from accelerated kinase activation caused by mutations in the activation loop. Strategies to overcome the loss of drug sensitivity that represents a challenge currently facing the field and the emerging approaches to circumvent resistance are discussed.


Asunto(s)
Resistencia a Antineoplásicos , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Benzamidas , Sitios de Unión , Proteínas de Fusión bcr-abl/química , Humanos , Enlace de Hidrógeno , Mesilato de Imatinib , Indoles/farmacología , Mutación , Fosforilación , Piperazinas/farmacología , Estabilidad Proteica , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/química , Proteínas Proto-Oncogénicas c-kit/química , Pirimidinas/farmacología , Pirroles/farmacología , Transducción de Señal , Sunitinib
3.
Proc Natl Acad Sci U S A ; 106(5): 1542-7, 2009 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-19164557

RESUMEN

Most gastrointestinal stromal tumors (GISTs) exhibit aberrant activation of the receptor tyrosine kinase (RTK) KIT. The efficacy of the inhibitors imatinib mesylate and sunitinib malate in GIST patients has been linked to their inhibition of these mutant KIT proteins. However, patients on imatinib can acquire secondary KIT mutations that render the protein insensitive to the inhibitor. Sunitinib has shown efficacy against certain imatinib-resistant mutants, although a subset that resides in the activation loop, including D816H/V, remains resistant. Biochemical and structural studies were undertaken to determine the molecular basis of sunitinib resistance. Our results show that sunitinib targets the autoinhibited conformation of WT KIT and that the D816H mutant undergoes a shift in conformational equilibrium toward the active state. These findings provide a structural and enzymologic explanation for the resistance profile observed with the KIT inhibitors. Prospectively, they have implications for understanding oncogenic kinase mutants and for circumventing drug resistance.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Indoles/uso terapéutico , Mutación , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Pirroles/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/metabolismo , Antineoplásicos/metabolismo , Benzamidas , Tumores del Estroma Gastrointestinal/enzimología , Tumores del Estroma Gastrointestinal/genética , Humanos , Mesilato de Imatinib , Indoles/metabolismo , Fosforilación , Piperazinas/metabolismo , Pirimidinas/metabolismo , Pirroles/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Espectrometría de Fluorescencia , Sunitinib
6.
J Med Chem ; 48(22): 6779-82, 2005 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-16250635

RESUMEN

A series of oxamyl dipeptides were optimized for pan caspase inhibition, anti-apoptotic cellular activity and in vivo efficacy. This structure-activity relationship study focused on the P4 oxamides and warhead moieties. Primarily on the basis of in vitro data, inhibitors were selected for study in a murine model of alpha-Fas-induced liver injury. IDN-6556 (1) was further profiled in additional in vivo models and pharmacokinetic studies. This first-in-class caspase inhibitor is now the subject of two Phase II clinical trials, evaluating its safety and efficacy for use in liver disease.


Asunto(s)
Inhibidores de Caspasas , Hepatopatías/tratamiento farmacológico , Ácidos Pentanoicos/síntesis química , Adulto , Alanina Transaminasa/sangre , Animales , Apoptosis/efectos de los fármacos , Aspartato Aminotransferasas/sangre , Disponibilidad Biológica , Caspasa 3 , Colestasis/tratamiento farmacológico , Colestasis/patología , Ensayos Clínicos Fase I como Asunto , Semivida , Hepatitis C Crónica/tratamiento farmacológico , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Células Jurkat , Hígado/efectos de los fármacos , Hígado/patología , Hepatopatías/enzimología , Hepatopatías/etiología , Ratones , Ácidos Pentanoicos/química , Ácidos Pentanoicos/farmacología , Ratas , Relación Estructura-Actividad
7.
Nat Commun ; 6: 7645, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26134520

RESUMEN

SIRT1, the founding member of the mammalian family of seven NAD(+)-dependent sirtuins, is composed of 747 amino acids forming a catalytic domain and extended N- and C-terminal regions. We report the design and characterization of an engineered human SIRT1 construct (mini-hSIRT1) containing the minimal structural elements required for lysine deacetylation and catalytic activation by small molecule sirtuin-activating compounds (STACs). Using this construct, we solved the crystal structure of a mini-hSIRT1-STAC complex, which revealed the STAC-binding site within the N-terminal domain of hSIRT1. Together with hydrogen-deuterium exchange mass spectrometry (HDX-MS) and site-directed mutagenesis using full-length hSIRT1, these data establish a specific STAC-binding site and identify key intermolecular interactions with hSIRT1. The determination of the interface governing the binding of STACs with human SIRT1 facilitates greater understanding of STAC activation of this enzyme, which holds significant promise as a therapeutic target for multiple human diseases.


Asunto(s)
Lisina/metabolismo , Sirtuina 1/química , Secuencia de Aminoácidos , Sitios de Unión/genética , Dominio Catalítico/genética , Cristalización , Cristalografía por Rayos X , Medición de Intercambio de Deuterio , Escherichia coli , Vectores Genéticos , Humanos , Espectrometría de Masas , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Sirtuina 1/genética , Sirtuina 1/metabolismo , Transfección
8.
Assay Drug Dev Technol ; 1(4): 527-35, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15090249

RESUMEN

A simple diagnostic method for mechanistic analysis of reversible enzyme inhibitors is presented. The method involves simple experimentation to determine how the inhibition by a reversible inhibitor changes in response to the substrate concentration varied in the assay. Four types of inhibitors are categorized based on their kinetic characteristic: (1) competitive or mutually exclusive inhibitors that compete with the substrate for the enzyme; (2) noncompetitive inhibitors that are independent of the substrate for binding to the enzyme; (3) antagonistic inhibitors where the binding affinity of the inhibitor is partially reduced by the substrate binding to the enzyme; and (4) synergistic inhibitors where inhibitor binding is enhanced by substrate binding. An equation was derived for data fitting and subsequent determination of inhibitor binding mode. The method was evaluated in three model enzyme systems, i.e., PK, adenylate kinase, and LDH, with known inhibitors. The method was also used to characterize a large number of unknown Csp3 inhibitors identified from HTS of a compound library consisting of 120,000 distinct chemical entities, a field test that validated the utility of the method. Among 76 Csp3 inhibitors analyzed, 70 were found to be non-mutually exclusive inhibitors, suggesting the existence of an allosteric site(s) in Csp3 for effective inhibition. The implication of this observation is discussed.


Asunto(s)
Inhibidores Enzimáticos/análisis , Cinética , Métodos , Adenilato Quinasa/análisis , Adenilato Quinasa/antagonistas & inhibidores , Algoritmos , Animales , Sitios de Unión/efectos de los fármacos , Caspasa 3 , Inhibidores de Caspasas , Caspasas/análisis , Fosfatos de Dinucleósidos/farmacocinética , Interacciones Farmacológicas , Inhibidores Enzimáticos/clasificación , Inhibidores Enzimáticos/metabolismo , L-Lactato Deshidrogenasa/análisis , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Modelos Teóricos , Piruvato Quinasa/análisis , Piruvato Quinasa/antagonistas & inhibidores , Conejos , Especificidad por Sustrato/efectos de los fármacos , Porcinos
9.
Expert Opin Investig Drugs ; 22(6): 675-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23705633

RESUMEN

In this issue of Expert Opinion on Investigational Drugs, several protein kinases families and pathways underlying cancer and other diseases are reviewed and several small molecule inhibitors that are in clinical trials are further described. Highlights of these reviews and drug evaluations are summarized in this editorial.


Asunto(s)
Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/efectos de los fármacos , Animales , Ensayos Clínicos como Asunto , Diseño de Fármacos , Humanos , Neoplasias/enzimología , Neoplasias/patología , Proteínas Quinasas/metabolismo
10.
Biotechniques ; 48(4): 303-11, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20569207

RESUMEN

The cleavage of double-stranded RNA (dsRNA) molecules by Dicer is a critical step in silencing genes by the RNA induced silencing complex (RISC). The development of Dicer substrates as nucleic acid-based therapeutics brings about a need to rapidly evaluate chemically modified RNA molecules for their ability to be processed by Dicer. This study outlines a quantitative fluorescence quencher-based assay for studying the ability of Dicer substrates to be processed by Dicer. By using a dsRNA probe labeled with Cy5-Iowa Black RQ, a panel of unlabeled test substrates can be rapidly assessed in heterologous competition assays without the need for electrophoresis or radiolabeling. This assay was piloted by evaluation of 196 unlabeled 27-mer Dicer substrates with various overhang structures in a purified Dicer enzyme assay system. Results indicate that Dicer has no preference for the sequence of RNA in the main double-stranded region of the substrate. However, a preference for Dicer substrate RNAs (D-siRNAs) containing purine/purine 3' overhang nucleotides was observed. These results demonstrate that the method is useful for studying the effects of modified nucleic acids in addition to rapidly accessing the influence of potential regulatory factors on Dicer processing.


Asunto(s)
Colorantes Fluorescentes/química , ARN Bicatenario/química , ARN Interferente Pequeño/química , Ribonucleasa III/metabolismo , Secuencia de Bases , Unión Competitiva , Carbocianinas/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Cinética , Datos de Secuencia Molecular , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/metabolismo , Espectrometría de Fluorescencia , Especificidad por Sustrato
11.
Protein Sci ; 19(4): 703-15, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20095048

RESUMEN

Mutations of the receptor tyrosine kinase KIT are linked to certain cancers such as gastrointestinal stromal tumors (GISTs). Biophysical, biochemical, and structural studies have provided insight into the molecular basis of resistance to the KIT inhibitors, imatinib and sunitinib. Here, solution-phase hydrogen/deuterium exchange (HDX) and direct binding mass spectrometry experiments provide a link between static structure models and the dynamic equilibrium of the multiple states of KIT, supporting that sunitinib targets the autoinhibited conformation of WT-KIT. The D816H mutation shifts the KIT conformational equilibrium toward the activated state. The V560D mutant exhibits two low energy conformations: one is more flexible and resembles the D816H mutant shifted toward the activated conformation, and the other is less flexible and resembles the wild-type KIT in the autoinhibited conformation. This result correlates with the V560D mutant exhibiting a sensitivity to sunitinib that is less than for WT KIT but greater than for KIT D816H. These findings support the elucidation of the resistance mechanism for the KIT mutants.


Asunto(s)
Proteínas Proto-Oncogénicas c-kit/química , Proteínas Proto-Oncogénicas c-kit/metabolismo , Sustitución de Aminoácidos , Benzamidas , Sitios de Unión , Medición de Intercambio de Deuterio , Resistencia a Medicamentos , Mesilato de Imatinib , Indoles/química , Indoles/metabolismo , Espectrometría de Masas/métodos , Datos de Secuencia Molecular , Piperazinas/química , Piperazinas/metabolismo , Pirimidinas/química , Pirimidinas/metabolismo , Pirroles/química , Pirroles/metabolismo , Sunitinib
12.
J Biochem ; 147(4): 601-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20147452

RESUMEN

The activation of receptor tyrosine kinases (RTKs) is tightly regulated through a variety of mechanisms. Kinetic studies show that activation of c-Kit RTK occurs through an inter-molecular autophosphorylation. Phosphopeptide mapping of c-Kit reveals that 14-22 phosphates are added to each mol of wild-type (WT) c-Kit during the activation. Phosphorylation sites are found on the JM, kinase insert (KID), c-terminal domains and the activation loop (A-loop), but only the sites on the JM domain contribute to the kinase activation. The A-loop tyrosine (Y(823)) is not phosphorylated until very late in the activation (>90% completion), indicating that the A-loop phosphorylation is not required for c-Kit activation. A sunitinib-resistant mutant D816H that accelerates auto-activation by 184-fold shows no phosphorylation on the A-loop tyrosine after full activation. A loss-of-phosphorylation mutation Y823F remains fully competent in auto-activation. Similar to WT and D816H, the unactivated Y823F mutant binds sunitinib and imatinib with high affinity (K(D) = 5.9 nM). But unlike the WT and D816H where the activated enzymes lose the ability to bind the two drugs, activated Y823F binds the two inhibitors effectively. These observations suggest that the A-loop of activated Y823F remains flexible and can readily adopt unactivated conformations to accommodate DFG-out binders.


Asunto(s)
Antineoplásicos/metabolismo , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/metabolismo , Indoles/metabolismo , Fosfotirosina/fisiología , Dominios y Motivos de Interacción de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirroles/metabolismo , Sustitución de Aminoácidos , Benzamidas , Dominio Catalítico , Activación Enzimática , Humanos , Mesilato de Imatinib , Cinética , Microquímica/métodos , Modelos Biológicos , Proteínas Mutantes/química , Proteínas Mutantes/aislamiento & purificación , Proteínas Mutantes/metabolismo , Mapeo Peptídico , Fosforilación , Piperazinas/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/química , Proteínas Proto-Oncogénicas c-kit/genética , Pirimidinas/metabolismo , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Sunitinib
13.
Chem Biol Drug Des ; 74(6): 547-59, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19843080

RESUMEN

The design, synthesis and utility of fluorescence probes that bind to the DFG-out conformation of p38alpha kinase are described. Probes that demonstrate good affinity for p38alpha, have been identified and one of the probes, PF-04438255, has been successfully used in an high throughput screening (HTS) assay to identify two novel non-classical p38alpha inhibitors. In addition, a cascade activity assay was utilized to validate the selective binding of these non-classical kinase inhibitors to the unactive form of the enzyme.


Asunto(s)
Colorantes Fluorescentes/síntesis química , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/síntesis química , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacología , Ensayos Analíticos de Alto Rendimiento , Cinética , Proteína Quinasa 14 Activada por Mitógenos/química , Naftalenos/química , Naftalenos/farmacología , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/química , Pirazoles/farmacología , Relación Estructura-Actividad
14.
Biochemistry ; 44(13): 4971-6, 2005 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-15794635

RESUMEN

ATP or dATP is a required activator of Apaf-1 for formation of the Apoptosome and thereby activation of caspase-9 (Csp9) [Zou, H., Henzel, W. J., Liu, X., Lutschg, A., and Wang, X. (1997) Cell 90, 405-413]. Here we demonstrate that dATP or ATP may have an additional role in controlling Apaf-1-mediated Csp9 activation. In the presence of cytochrome c (CytC), dATP or ATP binds to Apaf-1 and triggers heptamerization of Apaf-1 leading to the activation of Csp9. At concentrations greater than 1 mM, dATP or ATP also functions as a negative regulator of apoptosis by binding to and inhibiting Csp9. The affinity labeling reagent, 3'-O-(5-fluoro-2,4-dinitrophenyl)-ATP (FDNP-ATP), was used to probe the binding of nucleotides to Csp9. Similar to ATP, but with a much more profound effect, FDNP-ATP binds to the full-length proCsp9 potently, with an IC(50) of approximately 5-11 nM. Neither ATP nor FDNP-ATP exhibits any effect on the prodomain-truncated enzyme DeltaproCsp9 or p18/p10. FDNP-ATP covalently labels proCsp9 with a stoichiometry of 1:1, resulting in DNP-ATP-proCsp9 that is incapable of forming a productive Apoptosome with Apaf-1. Activity assays show that ATP and dATP, but not ADP or AMP, bind to the processed Csp9 p35/p10. This nucleotide binding site might play an important and previously unrecognized role in regulating proCsp9 activation.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Apoptosis/fisiología , Caspasas/química , Caspasas/metabolismo , Nucleótidos de Desoxiadenina/metabolismo , Marcadores de Afinidad , Factor Apoptótico 1 Activador de Proteasas , Sitios de Unión , Caspasa 9 , Activación Enzimática , Células HeLa , Humanos , Técnicas In Vitro , Cinética , Proteínas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
15.
Proc Natl Acad Sci U S A ; 102(19): 6954-9, 2005 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-15863619

RESUMEN

We have previously demonstrated an increase in proapoptotic caspase-3 in the kidney of Han:SPRD rats with polycystic kidney disease (PKD). The aim of the present study was to determine the effect of caspase inhibition on tubular cell apoptosis and proliferation, cyst formation, and renal failure in the Han:SPRD rat model of PKD. Heterozygous (Cy/+) and littermate control (+/+) male rats were weaned at 3 weeks of age and then treated with the caspase inhibitor IDN-8050 (10 mg/kg per day) by means of an Alzet (Palo Alto, CA) minipump or vehicle [polyethylene glycol (PEG 300)] for 5 weeks. The two-kidney/total body weight ratio more than doubled in Cy/+ rats compared with +/+ rats. IDN-8050 significantly reduced the kidney enlargement by 44% and the cyst volume density by 29% in Cy/+ rats. Cy/+ rats with PKD have kidney failure as indicated by a significant increase in blood urea nitrogen. IDN-8050 significantly reduced the increase in blood urea nitrogen in the Cy/+ rats. The number of proliferating cell nuclear antigen-positive tubular cells and apoptotic tubular cells in non-cystic and cystic tubules was significantly reduced in IDN-8050-treated Cy/+ rats compared with vehicle-treated Cy/+ rats. On immunoblot, the active form of caspase-3 (20 kDa) was significantly decreased in IDN-8050-treated Cy/+ rats compared with vehicle-treated Cy/+ rats. In summary, in a rat model of PKD, caspase inhibition with IDN-8050 (i) decreases apoptosis and proliferation in cystic and noncystic tubules; (ii) inhibits renal enlargement and cystogenesis, and (iii) attenuates the loss of kidney function.


Asunto(s)
Apoptosis , Inhibidores de Caspasas , Inhibidores Enzimáticos/farmacología , Animales , Animales Modificados Genéticamente , Nitrógeno de la Urea Sanguínea , Calpaína/metabolismo , Caspasa 3 , Caspasas/metabolismo , Catepsina B/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Heterocigoto , Immunoblotting , Etiquetado Corte-Fin in Situ , Concentración 50 Inhibidora , Riñón/metabolismo , Masculino , Ratones , Ratones Transgénicos , Enfermedades Renales Poliquísticas/patología , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas
17.
Biochemistry ; 42(14): 4151-60, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12680769

RESUMEN

Amino acid sequences of caspases 1, 3, 7, and 8 were aligned with their published three-dimensional (3D) structures. The resultant alignment was used as a template to compare the primary structures of caspases 2, 4-6, and 9-11 to build 3D homology models. The structural models were subsequently refined and validated using structure-activity relationship data obtained from an array of substrate-like inhibitors. All caspases were shown to have identical S1 and catalytic dyad architecture but diverse S2-S4 structures. S2 pockets of these 11 caspases can be briefly categorized into two groups: Csp3, -6, and -7 as one and Csp1, -2, -4, -5, -8, -9, -10, and -11 as the other. S2 pockets of Csp3, -6, and -7 are smaller than those of the other eight caspases, and are limited to binding small P2 residues such as Ala and Val. At the S3 site, the presence of a conserved Arg in all caspases suggests that Glu would be a universally preferred P3 residue. Csp8 and Csp9 have an additional Arg in this pocket that can further enhance the binding of a P3 Glu, whereas Csp2 has a Glu adjacent to the conserved Arg. As such, Csp2 is the only caspase that can accommodate both positively and negatively charged P3. At S4, Csp1, -4, -5, and -11 are closely related with respect to their structures and binder preferences; all have a large hydrophobic pocket and prefer large hydrophobic residues such as Trp. S4 of Csp2, -3, and -7 represents an opposite group with a conformation that is highly specific in binding an Asp. The S4 structures of Csp6, -8, -9, and -10 appear to be hybrids of the two extremes, and have little specificity for any P4. Information revealed from this work provides a guide for designing potent caspase inhibitors with desirable specificity.


Asunto(s)
Caspasas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Caspasas/química , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
18.
J Mol Recognit ; 16(3): 121-4, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12833566

RESUMEN

Caspases are cysteine proteases that play a critical role in the initiation and regulation of apoptosis. These enzymes act in a cascade to promote cell death through proteolytic cleavage of intracellular proteins. Since activation of apoptosis is implicated in human diseases such as cancer and neurodegenerative disorders, caspases are targets for drugs designed to modulate their action. Active caspases are heterodimeric enzymes with two symmetrically arranged active sites at opposite ends of the molecule. A number of crystal structures of caspases with peptides or proteins bound at the active sites have defined the mechanism of action of these enzymes, but molecular information about the active sites before substrate engagement has been lacking. As part of a study of peptidyl inhibitors of caspase-3, we crystallized a complex where the inhibitor did not bind in the active site. Here we present the crystal structure of the unoccupied substrate-binding site of caspase-3. No large conformational differences were apparent when this site was compared with that in enzyme-inhibitor complexes. Instead, the 1.9 A structure reveals critical side chain movements in a hydrophobic pocket in the active site. Notably, the side chain of tyrosine204 is rotated by approximately 90 degrees so that the phenol group occupies the S2 subsite in the active site. Thus, binding of substrate or inhibitors is impeded unless rotation of this side chain opens the area. The positions of these side chains may have important implications for the directed design of inhibitors of caspase-3 or caspase-7.


Asunto(s)
Caspasas/química , Caspasas/metabolismo , Apoptosis , Sitios de Unión , Caspasa 3 , Línea Celular , Cristalización , Cristalografía por Rayos X , Humanos , Ligandos , Modelos Moleculares , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas/antagonistas & inhibidores , Proteína Inhibidora de la Apoptosis Ligada a X
19.
J Biol Chem ; 278(10): 8091-8, 2003 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-12506111

RESUMEN

The apoptosome is a multiprotein complex comprising Apaf-1, cytochrome c, and caspase-9 that functions to activate caspase-3 downstream of mitochondria in response to apoptotic signals. Binding of cytochrome c and dATP to Apaf-1 in the cytosol leads to the assembly of a heptameric complex in which each Apaf-1 subunit is bound noncovalently to a procaspase-9 subunit via their respective CARD domains. Assembly of the apoptosome results in the proteolytic cleavage of procaspase-9 at the cleavage site PEPD(315) to yield the large (p35) and small (p12) caspase-9 subunits. In addition to the PEPD site, caspase-9 contains a caspase-3 cleavage site (DQLD(330)), which when cleaved, produces a smaller p10 subunit in which the NH(2)-terminal 15 amino acids of p12, including the XIAP BIR3 binding motif, are removed. Using purified proteins in a reconstituted reaction in vitro, we have assessed the relative impact of Asp(315) and Asp(330) cleavage on caspase-9 activity within the apoptosome. In addition, we characterized the effect of caspase-3 feedback cleavage of caspase-9 on the rate of caspase-3 activation, and the potential ramifications of Asp(330) cleavage on XIAP-mediated inhibition of the apoptosome. We have found that cleavage of procaspase-9 at Asp(330) to generate p35, p10 or p37, p10 forms resulted in a significant increase (up to 8-fold) in apoptosome activity compared with p35/p12. The significance of this increase was demonstrated by the near complete loss of apoptosome-mediated caspase-3 activity when a point mutant (D330A) of procaspase-9 was substituted for wild-type procaspase-9 in the apoptosome. In addition, cleavage at Asp(330) exposed a novel p10 NH(2)-terminal peptide motif (AISS) that retained the ability to mediate XIAP inhibition of caspase-9. Thus, whereas feedback cleavage of caspase-9 by caspase-3 significantly increases the activity of the apoptosome, it does little to attenuate its sensitivity to inhibition by XIAP.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Proteínas/metabolismo , Secuencia de Aminoácidos , Animales , Factor Apoptótico 1 Activador de Proteasas , Caspasa 3 , Caspasa 9 , Inhibidores de Caspasas , Caspasas/química , Grupo Citocromo c/metabolismo , Activación Enzimática , Humanos , Hidrólisis , Cinética , Datos de Secuencia Molecular , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Proteína Inhibidora de la Apoptosis Ligada a X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA