Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Org Chem ; 66(10): 3253-64, 2001 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-11348105

RESUMEN

Protein prenylation is a posttranslational lipid modification in which C(15) and C(20) isoprenoid units are linked to specific protein-derived cysteine residues through a thioether linkage. This process is catalyzed by a class of enzymes called prenyltransferases that are being intensively studied due to the finding that Ras protein is farnesylated coupled with the observation that mutant forms of Ras are implicated in a variety of human cancers. Inhibition of this posttranslational modification may serve as a possible cancer chemotherapy. Here, the syntheses of two new farnesyl diphosphate (FPP) analogues containing photoactive benzophenone groups are described. Each of these compounds was prepared in six steps from dimethylallyl alcohol. Substrate studies, inhibition kinetics, photoinactivation studies, and photolabeling experiments are also included; these experiments were performed with a number of protein prenyltransferases from different sources. A X-ray crystal structure of one of these analogues bound to rat farnesyltransferase illustrates that they are good substrate mimics. Of particular importance, these new analogues can be enzymatically incorporated into Ras-based peptide substrates allowing the preparation of molecules with photoactive isoprenoids that may serve as valuable probes for the study of prenylation function. Photoaffinity labeling of human protein geranylgeranyltransferase with (32)P-labeled forms of these analogues suggests that the C-10 locus of bound geranylgeranyl diphosphate (GGPP) is in close proximity to residues from the beta-subunit of this enzyme. These results clearly demonstrate the utility of these compounds as photoaffinity labeling analogues for the study of a variety of protein prenyltransferases and other enzymes that employ FPP or GGPP as their substrates.


Asunto(s)
Dimetilaliltranstransferasa/antagonistas & inhibidores , Fosfatos de Poliisoprenilo/síntesis química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Benzofenonas , Cristalografía por Rayos X , Dimetilaliltranstransferasa/metabolismo , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Éteres , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Radioisótopos de Fósforo , Fotoquímica , Fosfatos de Poliisoprenilo/química , Fosfatos de Poliisoprenilo/farmacocinética , Ratas , Sesquiterpenos , Relación Estructura-Actividad , Levaduras
2.
J Biol Chem ; 275(29): 21870-6, 2000 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-10749864

RESUMEN

Protein prenylation occurs in the protozoan that causes African sleeping sickness (Trypanosoma brucei), and the protein farnesyltransferase appears to be a good target for developing drugs. We have cloned the alpha- and beta-subunits of T. brucei protein farnesyltransferase (TB-PFT) using nucleic acid probes designed from partial amino acid sequences obtained from the enzyme purified from insect stage parasites. TB-PFT is expressed in both bloodstream and insect stage parasites. Enzymatically active TB-PFT was produced by heterologous expression in Escherichia coli. Compared with mammalian protein farnesyltransferases, TB-PFT contains a number of inserts of >25 residues in both subunits that reside on the surface of the enzyme in turns linking adjacent alpha-helices. Substrate specificity studies with a series of 20 peptides SSCALX (where X indicates a naturally occurring amino acid) show that the recombinant enzyme behaves identically to the native enzyme and displays distinct specificity compared with mammalian protein farnesyltransferase. TB-PFT prefers Gln and Met at the X position but not Ser, Thr, or Cys, which are good substrates for mammalian protein farnesyltransferase. A structural homology model of the active site of TB-PFT provides a basis for understanding structure-activity relations among substrates and CAAX mimetic inhibitors.


Asunto(s)
Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Trypanosoma brucei brucei/enzimología , Trypanosoma brucei brucei/genética , Transferasas Alquil y Aril/química , Secuencia de Aminoácidos , Animales , Clonación Molecular , Datos de Secuencia Molecular , Conformación Proteica , Ratas , Alineación de Secuencia , Análisis de Secuencia de Proteína , Especificidad por Sustrato
3.
Biochemistry ; 38(35): 11239-49, 1999 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-10471273

RESUMEN

Farnesyl protein transferase (FPT) is an alpha/beta heterodimeric zinc enzyme that catalyzes posttranslational farnesylation of many key cellular regulatory proteins, including oncogenic Ras. On the basis of the recently reported crystal structure of FPT complexed with a CVIM peptide and alpha-hydroxyfarnesylphosphonic acid, site-directed mutagenesis of the FPT active site was performed so key residues that are responsible for substrate binding and catalysis could be identified. Eight single mutants, including K164N alpha, Y166F alpha, Y166A alpha, Y200F alpha, H201A alpha, H248A beta, Y300F beta, and Y361F beta, and a double mutant, H248A beta/Y300F beta, were prepared. Steady-state kinetic analysis along with structural evidence indicated that residues Y200 alpha, H201 alpha, H248 beta, and Y361 beta are mainly involved in substrate binding. In addition, biochemical results confirm structural observations which show that residue Y166 alpha plays a key role in stabilizing the active site conformation of several FPT residues through cation-pi interactions. Two mutants, K164N alpha and Y300F beta, have moderately decreased catalytic constants (kcat). Pre-steady-state kinetic analysis of these mutants from rapid quench experiments showed that the chemical step rate constant was reduced by 41- and 30-fold, respectively. The product-releasing rate for each dropped approximately 10-fold. In pH-dependent kinetic studies, Y300F beta was observed to have both acidic and basic pKa values shifted 1 log unit from those of the wild-type enzyme, consistent with a possible role for Y300 beta as an acid-base catalyst. K164N alpha had a pKa shift from 6.0 to 5.3, which suggests it may function as a general acid. On the basis of these results along with structural evidence, a possible FPT reaction mechanism is proposed with both Y300 beta and K164 alpha playing key catalytic roles in enhancing the reactivity of the farnesyl diphosphate leaving group.


Asunto(s)
Transferasas Alquil y Aril/genética , Transferasas Alquil y Aril/metabolismo , Lisina/genética , Lisina/metabolismo , Tirosina/genética , Tirosina/metabolismo , Transferasas Alquil y Aril/química , Animales , Sitios de Unión/genética , Catálisis , Concentración de Iones de Hidrógeno , Cinética , Lisina/química , Modelos Químicos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Prenilación de Proteína/genética , Ratas , Tirosina/química
4.
J Med Chem ; 42(12): 2125-35, 1999 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-10377218

RESUMEN

Crystallographic and thermodynamic studies of farnesyl protein transferase (FPT) complexed with novel tricyclic inhibitors provide insights into the observed SAR for this unique class of nonpeptidic FPT inhibitors. The crystallographic structures reveal a binding pattern conserved across the mono-, di-, and trihalogen series. In the complexes, the tricycle spans the FPT active site cavity and interacts with both protein atoms and the isoprenoid portion of bound farnesyl diphosphate. An amide carbonyl, common to the tricyclic compounds described here, participates in a water-mediated hydrogen bond to the protein backbone. Ten high-resolution crystal structures of inhibitors complexed with FPT are reported. Included are crystallographic data for FPT complexed with SCH 66336, a compound currently undergoing clinical trials as an anticancer agent (SCH 66336, 4-[2-[4-(3,10-dibromo-8-chloro-6,11-dihydro-5H-benzo[5, 6]cyclohepta[1, 2-b]pyridin-11-yl)-1-piperidinyl]-2-oxoethyl]-1-piperidinecarbo xamide ). Thermodynamic binding parameters show favorable enthalpies of complex formation and small net entropic contributions as observed for 4-[2-[4-(3,10-dibromo-8-chloro-6,11-dihydro-11H-benzo[5, 6]cyclohepta[1, 2-b]pyridin-11-ylidene)-1-piperidinyl]-2-oxoethyl]pyridine N-oxide where DeltaH degrees bind = -12.5 kcal/mol and TDeltaS degrees bind = -1.5 kcal/mol.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Antineoplásicos/química , Óxidos N-Cíclicos/química , Inhibidores Enzimáticos/química , Compuestos Heterocíclicos con 3 Anillos/química , Piperidinas/química , Prenilación de Proteína , Piridinas/química , Sitios de Unión , Calorimetría , Cristalografía por Rayos X , Enlace de Hidrógeno , Modelos Moleculares , Termodinámica
5.
Protein Eng ; 12(4): 341-8, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10325405

RESUMEN

Protein farnesyltransferase (FPT) is a 97 000 Da heterodimeric enzyme that catalyzes post-translational farnesylation of many cellular regulatory proteins including p21 Ras. To facilitate the construction of site-directed mutants, a novel translationally coupled, two-cistron Escherichia coli expression system for rat FPT has been developed. This expression system enabled yields of >5 mg of purified protein per liter of E.coli culture to be obtained. The E.coli-derived FPT demonstrated an activity comparable to that of protein isolated from other sources. The reported expression system was used to construct three beta-subunit C-terminal truncation mutants, Delta5, Delta10 and Delta14, which were designed to eliminate a lattice interaction between the beta-subunit C-terminus of one molecule and the active site of a symmetry-related molecule. Steady-state kinetic analyses of these mutants showed that deletion up to 14 residues at the C-terminus did not reduce the value of kcat; however, Km values for both peptide and FPP increased 2-3-fold. A new crystalline form of FPT was obtained for the Delta10 C-terminal mutant grown in the presence of the substrate analogs acetyl-Cys-Val-Ile-Met-COOH peptide and alpha-hydroxyfarnesylphosphonic acid. The crystals diffract to beyond 2.0 A resolution. The refined structure clearly shows that both substrate analogs adopt extended conformations within the FPT active site cavity.


Asunto(s)
Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/aislamiento & purificación , Transferasas Alquil y Aril/metabolismo , Transferasas Alquil y Aril/genética , Animales , Cristalografía por Rayos X , Cartilla de ADN , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Escherichia coli/metabolismo , Farnesiltransferasa , Humanos , Cinética , Modelos Químicos , Mutagénesis , Plásmidos , Conformación Proteica , Ratas , Trombina/metabolismo
6.
Curr Opin Drug Discov Devel ; 2(5): 475-83, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19649974

RESUMEN

Emerging therapies for the treatment of cancer rely on detailed knowledge of cell growth mechanisms. It is hoped that mechanism-based inhibitors will prove more effective and exhibit fewer side effects than current treatments. Among the first, non-cytotoxic anticancer agents to enter clinical trials are the farnesyl protein transferase inhibitors, which prevent attachment of the farnesyl isoprenoid side chain to Ras. This post-translational modification is essential for the function of Ras, a protein exhibiting unregulated activity in nearly one-third of human cancers. Here structural studies of farnesyl protein transferase and strategies for discovery of its inhibitors are reviewed.

7.
Biochemistry ; 37(47): 16601-11, 1998 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-9843427

RESUMEN

The crystallographic structure of acetyl-Cys-Val-Ile-selenoMet-COOH and alpha-hydroxyfarnesylphosphonic acid (alphaHFP) complexed with rat farnesyl protein transferase (FPT) (space group P61, a = b = 174. 13 A, c = 69.71 A, alpha = beta = 90 degrees, gamma = 120 degrees, Rfactor = 21.8%, Rfree = 29.2%, 2.5 A resolution) is reported. In the ternary complex, the bound substrates are within van der Waals contact of each other and the FPT enzyme. alphaHFP binds in an extended conformation in the active-site cavity where positively charged side chains and solvent molecules interact with the phosphate moiety and aromatic side chains pack adjacent to the isoprenoid chain. The backbone of the bound CaaX peptide adopts an extended conformation, and the side chains interact with both FPT and alphaHFP. The cysteine sulfur of the bound peptide coordinates the active-site zinc. Overall, peptide binding and recognition appear to be dominated by side-chain interactions. Comparison of the structures of the ternary complex and unliganded FPT [Park, H., Boduluri, S., Moomaw, J., Casey, P., and Beese, L. (1997) Science 275, 1800-1804] shows that major rearrangements of several active site side chains occur upon substrate binding.


Asunto(s)
Transferasas Alquil y Aril/química , Oligopéptidos/química , Fosfatos de Poliisoprenilo/química , Transferasas Alquil y Aril/aislamiento & purificación , Transferasas Alquil y Aril/metabolismo , Animales , Sitios de Unión , Cristalización , Cristalografía por Rayos X , Farnesol/análogos & derivados , Farnesol/metabolismo , Humanos , Sustancias Macromoleculares , Modelos Moleculares , Oligopéptidos/metabolismo , Organofosfonatos/metabolismo , Fosfatos de Poliisoprenilo/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Sesquiterpenos , Especificidad por Sustrato
8.
Acta Crystallogr D Biol Crystallogr ; 54(Pt 6 Pt 2): 1207-15, 1998 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-10089499

RESUMEN

The X-ray crystallographic structure of [N-(3-phenylpropionyl)-N-(phenethyl)]-Gly-boroLys-OH (HPBK, Ki = 0. 42 nM, crystallographic R factor to 1.8 A resolution, 19.6%) complexed with human alpha-thrombin shows that the boron adopts a tetrahedral geometry and is covalently bonded to the active serine, Ser195. The HPBK phenethyl aromatic ring forms an edge-to-face interaction with the indole side chain of Trp215. Four HPBK analogs containing either electron-withdrawing or electron-donating substitutents at the 3' position of the phenethyl ring were synthesized in an attempt to modulate ligand affinity by inductive stabilization of the edge-to-face interaction. Refined crystallographic structures of the trifluoromethyl (Ki = 0.37 nM, crystallographic R factor to 2.0 A resolution = 18.7%), fluoro (Ki = 0.60; R factor to 2.3 A resolution = 18.4%), methoxy (Ki = 0.91 nM, R factor to 2.2 A resolution = 19.8%) and methyl (Ki = 0.20 nM, R factor to 2.5 A resolution = 16.9%) HPBK analogs complexed with thrombin revealed two binding modes for the closely related compounds. A less than 1.5-fold variation in affinity was observed for analogs (trifluoromethyl-HPBK and fluoro-HPBK) binding with the edge-to-face interaction. The slight inductive modulation is consistent with the overall weak nature of the edge-to-face interaction. Owing to an unexpected rotation of the phenethyl aromatic ring, the 3' substituent of two analogs, methoxy-HPBK and methyl-HPBK, made direct contact with the Trp215 indole side chain. Increased affinity of the 3' methyl analog is attributed to favorable interactions between the methyl group and the Trp215 indole ring. Differences in inhibitor, thrombin and solvent structure are discussed in detail. These results demonstrate the subtle interplay of weak forces that determine the equilibrium binding orientation of inhibitor, solvent and protein.


Asunto(s)
Anticoagulantes/química , Compuestos de Boro/química , Dipéptidos/química , Estructura Molecular , Trombina/antagonistas & inhibidores , Anticoagulantes/síntesis química , Anticoagulantes/metabolismo , Sitios de Unión , Compuestos de Boro/síntesis química , Compuestos de Boro/metabolismo , Compuestos de Boro/farmacología , Cristalografía por Rayos X , Dipéptidos/síntesis química , Dipéptidos/metabolismo , Dipéptidos/farmacología , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Electricidad Estática , Relación Estructura-Actividad , Especificidad por Sustrato , Trombina/química
9.
Acta Crystallogr D Biol Crystallogr ; 51(Pt 3): 337-41, 1995 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15299300

RESUMEN

TR1, a previously cloned gene for Crithidia fasciculata trypanothione reductase (TR), has been overexpressed in Escherichia coli strain SG5 to produce about 20 mg enzyme 1(-l) of culture. Since natural C. fasciculata TR is heterogeneous, this expression system provides an important source of homogeneous C. fasciculata TR for use in structural studies and drug design. Steady-state kinetic constants of the purified recombinant enzyme are K(m) = 56 micro M and k(cat) = 10 500 min(-1). Four crystal forms of TR1 were grown using this preparation. Synchrotron radiation was crucial to discover the high level of order present in crystal form IV, which diffracts to about 1.4 A resolution. To optimize growth and handling of form IV crystals, a novel crystallization setup called the 'plug drop' was developed.

10.
Proteins ; 20(3): 259-63, 1994 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-7892174

RESUMEN

A chimeric enzyme (GST121) of the human alpha-glutathione S-transferases GST1-1 and GST2-2, which has improved catalytic efficiency and thermostability from its wild-type parent proteins, has been crystallized in a space group that is isomorphous with that reported for crystals of GST1-1. However, a single-site (G82R) mutant of GST121, which exhibits a significant reduction both in vitro and in vivo in protein thermostability, forms crystals that are not isomorphous with GST1-1. The mutant protein crystallizes in space group P2(1)2(1)2(1), with cell dimensions a = 49.5, b = 92.9, c = 115.9 A, and one dimer per asymmetric unit. Preliminary crystallographic results show that a mutation of the surface residue Gly 82 from a neutral to a charged residue causes new salt bridges to be formed among the GST dimers, suggesting that the G82R mutant might aggregate more readily than does GST121 in solution, resulting in a change of its solution properties.


Asunto(s)
Glutatión Transferasa/genética , Secuencia de Aminoácidos , Cristalización , Cristalografía por Rayos X , Glutatión Transferasa/química , Calor , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Desnaturalización Proteica , Proteínas Recombinantes de Fusión/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...