Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(10)2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38791363

RESUMO

Protein farnesylation is a post-translational modification where a 15-carbon farnesyl isoprenoid is appended to the C-terminal end of a protein by farnesyltransferase (FTase). This process often causes proteins to associate with the membrane and participate in signal transduction pathways. The most common substrates of FTase are proteins that have C-terminal tetrapeptide CaaX box sequences where the cysteine is the site of modification. However, recent work has shown that five amino acid sequences can also be recognized, including the pentapeptides CMIIM and CSLMQ. In this work, peptide libraries were initially used to systematically vary the residues in those two parental sequences using an assay based on Matrix Assisted Laser Desorption Ionization-Mass Spectrometry (MALDI-MS). In addition, 192 pentapeptide sequences from the human proteome were screened using that assay to discover additional extended CaaaX-box motifs. Selected hits from that screening effort were rescreened using an in vivo yeast reporter protein assay. The X-ray crystal structure of CMIIM bound to FTase was also solved, showing that the C-terminal tripeptide of that sequence interacted with the enzyme in a similar manner as the C-terminal tripeptide of CVVM, suggesting that the tripeptide comprises a common structural element for substrate recognition in both tetrapeptide and pentapeptide sequences. Molecular dynamics simulation of CMIIM bound to FTase further shed light on the molecular interactions involved, showing that a putative catalytically competent Zn(II)-thiolate species was able to form. Bioinformatic predictions of tetrapeptide (CaaX-box) reactivity correlated well with the reactivity of pentapeptides obtained from in vivo analysis, reinforcing the importance of the C-terminal tripeptide motif. This analysis provides a structural framework for understanding the reactivity of extended CaaaX-box motifs and a method that may be useful for predicting the reactivity of additional FTase substrates bearing CaaaX-box sequences.


Assuntos
Biologia Computacional , Biblioteca de Peptídeos , Humanos , Biologia Computacional/métodos , Especificidade por Substrato , Farnesiltranstransferase/metabolismo , Farnesiltranstransferase/química , Oligopeptídeos/química , Oligopeptídeos/metabolismo , Sequência de Aminoácidos , Cristalografia por Raios X , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Ligação Proteica
2.
Protein Sci ; 23(3): 289-301, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24347326

RESUMO

Species of the fungal genus Aspergillus are significant human and agricultural pathogens that are often refractory to existing antifungal treatments. Protein farnesyltransferase (FTase), a critical enzyme in eukaryotes, is an attractive potential target for antifungal drug discovery. We report high-resolution structures of A. fumigatus FTase (AfFTase) in complex with substrates and inhibitors. Comparison of structures with farnesyldiphosphate (FPP) bound in the absence or presence of peptide substrate, corresponding to successive steps in ordered substrate binding, revealed that the second substrate-binding step is accompanied by motions of a loop in the catalytic site. Re-examination of other FTase structures showed that this motion is conserved. The substrate- and product-binding clefts in the AfFTase active site are wider than in human FTase (hFTase). Widening is a consequence of small shifts in the α-helices that comprise the majority of the FTase structure, which in turn arise from sequence variation in the hydrophobic core of the protein. These structural effects are key features that distinguish fungal FTases from hFTase. Their variation results in differences in steady-state enzyme kinetics and inhibitor interactions and presents opportunities for developing selective anti-fungal drugs by exploiting size differences in the active sites. We illustrate the latter by comparing the interaction of ED5 and Tipifarnib with hFTase and AfFTase. In AfFTase, the wider groove enables ED5 to bind in the presence of FPP, whereas in hFTase it binds only in the absence of substrate. Tipifarnib binds similarly to both enzymes but makes less extensive contacts in AfFTase with consequently weaker binding.


Assuntos
Antifúngicos/farmacocinética , Aspergillus fumigatus/metabolismo , Farnesiltranstransferase/química , Farnesiltranstransferase/metabolismo , Peptídeos/química , Aspergillus fumigatus/química , Domínio Catalítico , Cristalografia por Raios X , Desenho de Fármacos , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Humanos , Peptídeos/antagonistas & inibidores , Fosfatos de Poli-Isoprenil/antagonistas & inibidores , Fosfatos de Poli-Isoprenil/química , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Quinolonas/farmacocinética , Sesquiterpenos/antagonistas & inibidores , Sesquiterpenos/química , Sulfonamidas/farmacocinética , Benzenossulfonamidas
3.
Artigo em Inglês | MEDLINE | ID: mdl-21821902

RESUMO

MutSß is a eukaryotic mismatch repair protein that preferentially targets extrahelical unpaired nucleotides and shares partial functional redundancy with MutSα (MSH2-MSH6). Although mismatch recognition by MutSα has been shown to involve a conserved Phe-X-Glu motif, little is known about the lesion-binding mechanism of MutSß. Combined MSH3/MSH6 deficiency triggers a strong predisposition to cancer in mice and defects in msh2 and msh6 account for roughly half of hereditary nonpolyposis colorectal cancer mutations. These three MutS homologs are also believed to play a role in trinucleotide repeat instability, which is a hallmark of many neurodegenerative disorders. The baculovirus overexpression and purification of recombinant human MutSß and three truncation mutants are presented here. Binding assays with heteroduplex DNA were carried out for biochemical characterization. Crystallization and preliminary X-ray diffraction analysis of the protein bound to a heteroduplex DNA substrate are also reported.


Assuntos
Proteína 2 Homóloga a MutS/química , Cristalização , Cristalografia por Raios X , Humanos , Proteína 2 Homóloga a MutS/genética , Proteína 2 Homóloga a MutS/isolamento & purificação , Mutação , Ligação Proteica
4.
J Biol Chem ; 286(40): 35149-62, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-21816822

RESUMO

Cryptococcus neoformans is a fungal pathogen that causes life-threatening infections in immunocompromised individuals, including AIDS patients and transplant recipients. Few antifungals can treat C. neoformans infections, and drug resistance is increasing. Protein farnesyltransferase (FTase) catalyzes post-translational lipidation of key signal transduction proteins and is essential in C. neoformans. We present a multidisciplinary study validating C. neoformans FTase (CnFTase) as a drug target, showing that several anticancer FTase inhibitors with disparate scaffolds can inhibit C. neoformans and suggesting structure-based strategies for further optimization of these leads. Structural studies are an essential element for species-specific inhibitor development strategies by revealing similarities and differences between pathogen and host orthologs that can be exploited. We, therefore, present eight crystal structures of CnFTase that define the enzymatic reaction cycle, basis of ligand selection, and structurally divergent regions of the active site. Crystal structures of clinically important anticancer FTase inhibitors in complex with CnFTase reveal opportunities for optimization of selectivity for the fungal enzyme by modifying functional groups that interact with structurally diverse regions. A substrate-induced conformational change in CnFTase is observed as part of the reaction cycle, a feature that is mechanistically distinct from human FTase. Our combined structural and functional studies provide a framework for developing FTase inhibitors to treat invasive fungal infections.


Assuntos
Alquil e Aril Transferases/química , Cryptococcus neoformans/metabolismo , Antifúngicos/farmacologia , Clonagem Molecular , Cristalografia por Raios X/métodos , Desenho de Fármacos , Humanos , Ligantes , Modelos Químicos , Prenilação , Conformação Proteica , Processamento de Proteína Pós-Traducional , Transdução de Sinais , Especificidade por Substrato
5.
J Med Chem ; 53(19): 6867-88, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20822181

RESUMO

A potent class of anticancer, human farnesyltransferase (hFTase) inhibitors has been identified by "piggy-backing" on potent, antimalarial inhibitors of Plasmodium falciparum farnesyltransferase (PfFTase). On the basis of a 4-fold substituted ethylenediamine scaffold, the inhibitors are structurally simple and readily derivatized, facilitating the extensive structure-activity relationship (SAR) study reported herein. Our most potent inhibitor is compound 1f, which exhibited an in vitro hFTase IC(50) value of 25 nM and a whole cell H-Ras processing IC(50) value of 90 nM. Moreover, it is noteworthy that several of our inhibitors proved highly selective for hFTase (up to 333-fold) over the related prenyltransferase enzyme geranylgeranyltransferase-I (GGTase-I). A crystal structure of inhibitor 1a co-crystallized with farnesyl pyrophosphate (FPP) in the active site of rat FTase illustrates that the para-benzonitrile moiety of 1a is stabilized by a π-π stacking interaction with the Y361ß residue, suggesting a structural explanation for the observed importance of this component of our inhibitors.


Assuntos
Antineoplásicos/síntese química , Etilenodiaminas/síntese química , Farnesiltranstransferase/antagonistas & inibidores , Modelos Moleculares , Compostos de Anilina/síntese química , Compostos de Anilina/química , Compostos de Anilina/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Domínio Catalítico , Linhagem Celular , Cristalografia por Raios X , Desenho de Fármacos , Etilenodiaminas/química , Etilenodiaminas/farmacologia , Humanos , Estrutura Molecular , Nitrilas/síntese química , Nitrilas/química , Nitrilas/farmacologia , Plasmodium falciparum/enzimologia , Ligação Proteica , Ratos , Relação Estrutura-Atividade , Sulfonamidas/síntese química , Sulfonamidas/química , Sulfonamidas/farmacologia
6.
Chem Biol ; 16(2): 181-92, 2009 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-19246009

RESUMO

Protein farnesyltransferase (FTase) catalyzes an essential posttranslational lipid modification of more than 60 proteins involved in intracellular signal transduction networks. FTase inhibitors have emerged as a significant target for development of anticancer therapeutics and, more recently, for the treatment of parasitic diseases caused by protozoan pathogens, including malaria (Plasmodium falciparum). We present the X-ray crystallographic structures of complexes of mammalian FTase with five inhibitors based on an ethylenediamine scaffold, two of which exhibit over 1000-fold selective inhibition of P. falciparum FTase. These structures reveal the dominant determinants in both the inhibitor and enzyme that control binding and selectivity. Comparison to a homology model constructed for the P. falciparum FTase suggests opportunities for further improving selectivity of a new generation of antimalarial inhibitors.


Assuntos
Antimaláricos/química , Antineoplásicos/química , Inibidores Enzimáticos/química , Farnesiltranstransferase/antagonistas & inibidores , Farnesiltranstransferase/química , Animais , Antimaláricos/metabolismo , Antineoplásicos/metabolismo , Linhagem Celular Tumoral , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/metabolismo , Etilenodiaminas/química , Farnesiltranstransferase/metabolismo , Humanos , Plasmodium falciparum/enzimologia , Ligação Proteica , Conformação Proteica , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Ratos , Homologia Estrutural de Proteína , Relação Estrutura-Atividade , Especificidade por Substrato
7.
Chem Biol Drug Des ; 72(3): 171-81, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18844669

RESUMO

Originally designed to block the prenylation of oncogenic Ras, inhibitors of protein farnesyltransferase currently in preclinical and clinical trials are showing efficacy in cancers with normal Ras. Blocking protein prenylation has also shown promise in the treatment of malaria, Chagas disease and progeria syndrome. A better understanding of the mechanism, targets and in vivo consequences of protein prenylation are needed to elucidate the mode of action of current PFTase (Protein Farnesyltransferase) inhibitors and to create more potent and selective compounds. Caged enzyme substrates are useful tools for understanding enzyme mechanism and biological function. Reported here is the synthesis and characterization of caged substrates of PFTase. The caged isoprenoid diphosphates are poor substrates prior to photolysis. The caged CAAX peptide is a true catalytically caged substrate of PFTase in that it is to not a substrate, yet is able to bind to the enzyme as established by inhibition studies and X-ray crystallography. Irradiation of the caged molecules with 350 nm light readily releases their cognate substrate and their photolysis products are benign. These properties highlight the utility of those analogs towards a variety of in vitro and in vivo applications.


Assuntos
Dimetilaliltranstransferase/metabolismo , Prenilação de Proteína/efeitos dos fármacos , Alquil e Aril Transferases/metabolismo , Cristalografia por Raios X , Inibidores Enzimáticos/metabolismo , Farnesiltranstransferase/metabolismo , Humanos , Peptídeos/metabolismo , Fosfatos de Poli-Isoprenil/química , Especificidade por Substrato/efeitos dos fármacos
8.
J Biol Chem ; 283(46): 31933-40, 2008 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-18713740

RESUMO

Protein geranylgeranyltransferase-I (GGTase-I) catalyzes the transfer of a 20-carbon isoprenoid lipid to the sulfur of a cysteine residue located near the C terminus of numerous cellular proteins, including members of the Rho superfamily of small GTPases and other essential signal transduction proteins. In humans, GGTase-I and the homologous protein farnesyltransferase (FTase) are targets of anticancer therapeutics because of the role small GTPases play in oncogenesis. Protein prenyltransferases are also essential for many fungal and protozoan pathogens that infect humans, and have therefore become important targets for treating infectious diseases. Candida albicans, a causative agent of systemic fungal infections in immunocompromised individuals, is one pathogen for which protein prenylation is essential for survival. Here we present the crystal structure of GGTase-I from C. albicans (CaGGTase-I) in complex with its cognate lipid substrate, geranylgeranylpyrophosphate. This structure provides a high-resolution picture of a non-mammalian protein prenyltransferase. There are significant variations between species in critical areas of the active site, including the isoprenoid-binding pocket, as well as the putative product exit groove. These differences indicate the regions where specific protein prenyltransferase inhibitors with antifungal activity can be designed.


Assuntos
Alquil e Aril Transferases/química , Alquil e Aril Transferases/metabolismo , Candida albicans/enzimologia , Metabolismo dos Lipídeos , Alquil e Aril Transferases/genética , Sítios de Ligação , Candida albicans/genética , Candida albicans/patogenicidade , Cristalografia por Raios X , Humanos , Modelos Moleculares , Ligação Proteica , Prenilação de Proteína , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Especificidade por Substrato
9.
J Biol Chem ; 283(47): 32812-20, 2008 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-18723845

RESUMO

Several bacterial solute transport mechanisms involve members of the periplasmic binding protein (PBP) superfamily that bind and deliver ligand to integral membrane transport proteins in the ATP-binding cassette, tripartite tricarboxylate transporter, or tripartite ATP-independent (TRAP) families. PBPs involved in ATP-binding cassette transport systems have been well characterized, but only a few PBPs involved in TRAP transport have been studied. We have measured the thermal stability, determined the oligomerization state by small angle x-ray scattering, and solved the x-ray crystal structure to 1.9 A resolution of a TRAP-PBP (open reading frame tm0322) from the hyperthermophilic bacterium Thermotoga maritima (TM0322). The overall fold of TM0322 is similar to other TRAP transport related PBPs, although the structural similarity of backbone atoms (2.5-3.1 A root mean square deviation) is unusually low for PBPs within the same group. Individual monomers within the tetrameric asymmetric unit of TM0322 exhibit high root mean square deviation (0.9 A) to each other as a consequence of conformational heterogeneity in their binding pockets. The gel filtration elution profile and the small angle x-ray scattering analysis indicate that TM0322 assembles as dimers in solution that in turn assemble into a dimer of dimers in the crystallographic asymmetric unit. Tetramerization has been previously observed in another TRAP-PBP (the Rhodobacter sphaeroides alpha-keto acid-binding protein) where quaternary structure formation is postulated to be an important requisite for the transmembrane transport process.


Assuntos
Trifosfato de Adenosina/química , Transporte Biológico , Clonagem Molecular , Cristalografia por Raios X , Ligantes , Proteínas de Membrana Transportadoras/química , Modelos Moleculares , Modelos Estatísticos , Conformação Molecular , Periplasma/metabolismo , Proteínas Periplásmicas de Ligação/química , Ligação Proteica , Thermotoga maritima/química
10.
J Biol Chem ; 283(19): 13310-9, 2008 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-18326858

RESUMO

We have examined the interaction parameters, conformation, and functional significance of the human MutSalpha(.) proliferating cell nuclear antigen (PCNA) complex in mismatch repair. The two proteins associate with a 1:1 stoichiometry and a K(D) of 0.7 microm in the absence or presence of heteroduplex DNA. PCNA does not influence the affinity of MutSalpha for a mismatch, and mismatch-bound MutSalpha binds PCNA. Small angle x-ray scattering studies have established the molecular parameters of the complex, which are consistent with an elongated conformation in which the two proteins associate in an end-to-end fashion in a manner that does not involve an extended unstructured tether, as has been proposed for yeast MutSalpha and PCNA ( Shell, S. S., Putnam, C. D., and Kolodner, R. D. (2007) Mol. Cell 26, 565-578 ). MutSalpha variants lacking the PCNA interaction motif are functional in 3'- or 5'-directed mismatch-provoked excision, but display a partial defect in 5'-directed mismatch repair. This finding is consistent with the modest mutability conferred by inactivation of the MutSalpha PCNA interaction motif and suggests that interaction of the replication clamp with other repair protein(s) accounts for the essential role of PCNA in MutSalpha-dependent mismatch repair.


Assuntos
Reparo de Erro de Pareamento de DNA , Proteínas de Ligação a DNA/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Antígeno Nuclear de Célula em Proliferação/metabolismo , Sequência de Aminoácidos , Fenômenos Biofísicos , Biofísica , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Dimerização , Células HeLa , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Proteína 2 Homóloga a MutS/química , Proteína 2 Homóloga a MutS/genética , Antígeno Nuclear de Célula em Proliferação/química , Ligação Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
11.
Protein Sci ; 16(10): 2240-50, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17766373

RESUMO

We report the design and engineering of a robust, reagentless fluorescent glucose biosensor based on the periplasmic glucose-binding protein obtained from Thermotoga maritima (tmGBP). The gene for this protein was cloned from genomic DNA and overexpressed in Escherichia coli, the identity of its cognate sugar was confirmed, ligand binding was studied, and the structure of its glucose complex was solved to 1.7 Angstrom resolution by X-ray crystallography. TmGBP is specific for glucose and exhibits high thermostability (midpoint of thermal denaturation is 119 +/- 1 degrees C and 144 +/- 2 degrees C in the absence and presence of 1 mM glucose, respectively). A series of fluorescent conjugates was constructed by coupling single, environmentally sensitive fluorophores to unique cysteines introduced by site-specific mutagenesis at positions predicted to be responsive to ligand-induced conformational changes based on the structure. These conjugates were screened to identify engineered tmGBPs that function as reagentless fluorescent glucose biosensors. The Y13C*Cy5 conjugate is bright, gives a large response to glucose over concentration ranges appropriate for in vivo monitoring of blood glucose levels (1-30 mM), and can be immobilized in an orientation-specific manner in microtiter plates to give a reversible response to glucose. The immobilized protein retains its response after long-term storage at room temperature.


Assuntos
Proteínas de Bactérias/química , Proteínas de Transporte/química , Corantes Fluorescentes/química , Glucose/análise , Thermotoga maritima , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Técnicas Biossensoriais , Carbocianinas/química , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Clonagem Molecular , Cristalografia por Raios X , Glucose/química , Glucose/metabolismo , Ligação de Hidrogênio , Ligantes , Modelos Moleculares , Mutagênese Sítio-Dirigida , Thermotoga maritima/genética
12.
Nucleic Acids Res ; 35(13): 4275-88, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17576677

RESUMO

We have investigated how a benzo[a]pyrene-derived N2-dG adduct, 10S(+)-trans-anti-[BP]-N2-dG ([BP]G*), is processed in a well-characterized Pol I family model replicative DNA polymerase, Bacillus fragment (BF). Experimental results are presented that reveal relatively facile nucleotide incorporation opposite the lesion, but very inefficient further extension. Computational studies follow the possible bypass of [BP]G* through the pre-insertion, insertion and post-insertion sites as BF alternates between open and closed conformations. With dG* in the normal B-DNA anti conformation, BP seriously disturbs the polymerase structure, positioning itself either deeply in the pre-insertion site or on the crowded evolving minor groove side of the modified template, consistent with a polymerase-blocking conformation. With dG* in the less prevalent syn conformation, BP causes less distortion: it is either out of the pre-insertion site or in the major groove open pocket of the polymerase. Thus, the syn conformation can account for the observed relatively easy incorporation of nucleotides, with mutagenic purines favored, opposite the [BP]G* adduct. However, with the lesion in the BF post-insertion site, more serious distortions caused by the adduct even in the syn conformation explain the very inefficient extension observed experimentally. In vivo, a switch to a potentially error-prone bypass polymerase likely dominates translesion bypass.


Assuntos
Benzopirenos/química , Carcinógenos/química , Adutos de DNA/química , DNA Polimerase I/metabolismo , Replicação do DNA , Desoxiguanosina/análogos & derivados , Sítios de Ligação , Biologia Computacional , DNA/biossíntese , DNA/química , DNA Polimerase I/química , Desoxiguanosina/química , Ligação de Hidrogênio , Modelos Moleculares , Conformação de Ácido Nucleico , Nucleotídeos/química , Nucleotídeos/metabolismo
13.
Mol Cell ; 26(4): 579-92, 2007 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-17531815

RESUMO

Mismatch repair (MMR) ensures the fidelity of DNA replication, initiates the cellular response to certain classes of DNA damage, and has been implicated in the generation of immune diversity. Each of these functions depends on MutSalpha (MSH2*MSH6 heterodimer). Inactivation of this protein complex is responsible for tumor development in about half of known hereditary nonpolyposis colorectal cancer kindreds and also occurs in sporadic tumors in a variety of tissues. Here, we describe a series of crystal structures of human MutSalpha bound to different DNA substrates, each known to elicit one of the diverse biological responses of the MMR pathway. All lesions are recognized in a similar manner, indicating that diversity of MutSalpha-dependent responses to DNA lesions is generated in events downstream of this lesion recognition step. This study also allows rigorous mapping of cancer-causing mutations and furthermore suggests structural pathways for allosteric communication between different regions within the heterodimer.


Assuntos
Pareamento Incorreto de Bases , Proteína MutS de Ligação de DNA com Erro de Pareamento/química , Difosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Neoplasias do Colo/genética , Sequência Conservada , Replicação do DNA , Humanos , Magnésio/metabolismo , Modelos Moleculares , Proteína MutS de Ligação de DNA com Erro de Pareamento/genética , Proteína MutS de Ligação de DNA com Erro de Pareamento/metabolismo , Conformação Proteica , Estrutura Secundária de Proteína , Alinhamento de Sequência
14.
Proc Natl Acad Sci U S A ; 103(52): 19701-6, 2006 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-17179038

RESUMO

Methylating agents are widespread environmental carcinogens that generate a broad spectrum of DNA damage. Methylation at the guanine O(6) position confers the greatest mutagenic and carcinogenic potential. DNA polymerases insert cytosine and thymine with similar efficiency opposite O(6)-methyl-guanine (O6MeG). We combined pre-steady-state kinetic analysis and a series of nine x-ray crystal structures to contrast the reaction pathways of accurate and mutagenic replication of O6MeG in a high-fidelity DNA polymerase from Bacillus stearothermophilus. Polymerases achieve substrate specificity by selecting for nucleotides with shape and hydrogen-bonding patterns that complement a canonical DNA template. Our structures reveal that both thymine and cytosine O6MeG base pairs evade proofreading by mimicking the essential molecular features of canonical substrates. The steric mimicry depends on stabilization of a rare cytosine tautomer in C.O6MeG-polymerase complexes. An unusual electrostatic interaction between O-methyl protons and a thymine carbonyl oxygen helps stabilize T.O6MeG pairs bound to DNA polymerase. Because DNA methylators constitute an important class of chemotherapeutic agents, the molecular mechanisms of replication of these DNA lesions are important for our understanding of both the genesis and treatment of cancer.


Assuntos
Guanina/análogos & derivados , Sítios de Ligação , Cristalografia por Raios X , DNA/química , DNA/genética , DNA/metabolismo , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/metabolismo , Guanina/química , Guanina/metabolismo , Cinética , Modelos Moleculares , Mutação/genética , Estrutura Terciária de Proteína
15.
Biochemistry ; 45(32): 9746-55, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16893176

RESUMO

Posttranslational modifications are essential for the proper function of a number of proteins in the cell. One such modification, the covalent attachment of a single isoprenoid lipid (prenylation), is carried out by the CaaX prenyltransferases, protein farnesyltransferase (FTase) and protein geranylgeranyltransferase type-I (GGTase-I). Substrate proteins of these two enzymes are involved in a variety of cellular functions but are largely associated with signal transduction. These modified proteins include members of the Ras superfamily, heterotrimeric G-proteins, centromeric proteins, and a number of proteins involved in nuclear integrity. Although FTase and GGTase-I are highly homologous, they are quite selective for their substrates, particularly for their isoprenoid diphosphate substrates, FPP and GGPP, respectively. Here, we present both crystallographic and kinetic analyses of mutants designed to explore this isoprenoid specificity and demonstrate that this specificity is dependent upon two enzyme residues in the beta subunits of the enzymes, W102beta and Y365beta in FTase (T49beta and F324beta, respectively, in GGTase-I).


Assuntos
Alquil e Aril Transferases/química , Alquil e Aril Transferases/metabolismo , Farnesiltranstransferase/química , Farnesiltranstransferase/metabolismo , Alquil e Aril Transferases/genética , Sítios de Ligação , Cristalografia por Raios X , Farnesiltranstransferase/genética , Humanos , Modelos Moleculares , Mutação/genética , Peptídeos/química , Prenilação de Proteína , Especificidade por Substrato
16.
J Lipid Res ; 47(4): 681-99, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16477080

RESUMO

More than 100 proteins necessary for eukaryotic cell growth, differentiation, and morphology require posttranslational modification by the covalent attachment of an isoprenoid lipid (prenylation). Prenylated proteins include members of the Ras, Rab, and Rho families, lamins, CENPE and CENPF, and the gamma subunit of many small heterotrimeric G proteins. This modification is catalyzed by the protein prenyltransferases: protein farnesyltransferase (FTase), protein geranylgeranyltransferase type I (GGTase-I), and GGTase-II (or RabGGTase). In this review, we examine the structural biology of FTase and GGTase-I (the CaaX prenyltransferases) to establish a framework for understanding the molecular basis of substrate specificity and mechanism. These enzymes have been identified in a number of species, including mammals, fungi, plants, and protists. Prenyltransferase structures include complexes that represent the major steps along the reaction path, as well as a number of complexes with clinically relevant inhibitors. Such complexes may assist in the design of inhibitors that could lead to treatments for cancer, viral infection, and a number of deadly parasitic diseases.


Assuntos
Alquil e Aril Transferases , Alquil e Aril Transferases/química , Alquil e Aril Transferases/metabolismo , Animais , Sítios de Ligação , Ciclo Celular/fisiologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Modelos Moleculares , Estrutura Molecular , Peptídeos/química , Peptídeos/metabolismo , Fosfatos de Poli-Isoprenil/química , Fosfatos de Poli-Isoprenil/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Especificidade por Substrato , Zinco/metabolismo
17.
J Biol Chem ; 280(5): 3764-70, 2005 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-15548515

RESUMO

Of the carcinogens to which humans are most frequently exposed, the polycyclic aromatic hydrocarbon benzo[a]pyrene (BP) is one of the most ubiquitous. BP is a byproduct of grilled foods and tobacco and fuel combustion and has long been linked to various human cancers, particularly lung and skin. BP is metabolized to diol epoxides that covalently modify DNA bases to form bulky adducts that block DNA synthesis by replicative or high fidelity DNA polymerases. Here we present the structure of a high fidelity polymerase from a thermostable strain of Bacillus stearothermophilus (Bacillus fragment) bound to the most common BP-derived N2-guanine adduct base-paired with cytosine. The BP adduct adopts a conformation that places the polycyclic BP moiety in the nascent DNA minor groove and is the first structure of a minor groove adduct bound to a polymerase. Orientation of the BP moiety into the nascent DNA minor groove results in extensive disruption to the interactions between the adducted DNA duplex and the polymerase. The disruptions revealed by the structure of Bacillus fragment bound to a BP adduct provide a molecular basis for rationalizing the potent blocking effect on replication exerted by BP adducts.


Assuntos
Benzo(a)pireno/metabolismo , Replicação do DNA/fisiologia , DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/genética , Geobacillus stearothermophilus/enzimologia , Sítios de Ligação , Cristalografia , Adutos de DNA/química , Adutos de DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Geobacillus stearothermophilus/genética , Estrutura Terciária de Proteína
18.
J Mol Biol ; 343(2): 417-33, 2004 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-15451670

RESUMO

Post-translational modifications are essential for the proper function of many proteins in the cell. The attachment of an isoprenoid lipid (a process termed prenylation) by protein farnesyltransferase (FTase) or geranylgeranyltransferase type I (GGTase-I) is essential for the function of many signal transduction proteins involved in growth, differentiation, and oncogenesis. FTase and GGTase-I (also called the CaaX prenyltransferases) recognize protein substrates with a C-terminal tetrapeptide recognition motif called the Ca1a2X box. These enzymes possess distinct but overlapping protein substrate specificity that is determined primarily by the sequence identity of the Ca1a2X motif. To determine how the identity of the Ca1a2X motif residues and sequence upstream of this motif affect substrate binding, we have solved crystal structures of FTase and GGTase-I complexed with a total of eight cognate and cross-reactive substrate peptides, including those derived from the C termini of the oncoproteins K-Ras4B, H-Ras and TC21. These structures suggest that all peptide substrates adopt a common binding mode in the FTase and GGTase-I active site. Unexpectedly, while the X residue of the Ca1a2X motif binds in the same location for all GGTase-I substrates, the X residue of FTase substrates can bind in one of two different sites. Together, these structures outline a series of rules that govern substrate peptide selectivity; these rules were utilized to classify known protein substrates of CaaX prenyltransferases and to generate a list of hypothetical substrates within the human genome.


Assuntos
Alquil e Aril Transferases/química , Conformação Proteica , Alquil e Aril Transferases/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Peptídeos/química , Peptídeos/metabolismo , Ligação Proteica , Prenilação de Proteína , Especificidade por Substrato
19.
J Biol Chem ; 279(48): 50280-5, 2004 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-15385534

RESUMO

Aromatic amines have been studied for more than a half-century as model carcinogens representing a class of chemicals that form bulky adducts to the C8 position of guanine in DNA. Among these guanine adducts, the N-(2'-deoxyguanosin-8-yl)-aminofluorene (G-AF) and N-2-(2'-deoxyguanosin-8-yl)-acetylaminofluorene (G-AAF) derivatives are the best studied. Although G-AF and G-AAF differ by only an acetyl group, they exert different effects on DNA replication by replicative and high-fidelity DNA polymerases. Translesion synthesis of G-AF is achieved with high-fidelity polymerases, whereas replication of G-AAF requires specialized bypass polymerases. Here we have presented structures of G-AF as it undergoes one round of accurate replication by a high-fidelity DNA polymerase. Nucleotide incorporation opposite G-AF is achieved in solution and in the crystal, revealing how the polymerase accommodates and replicates past G-AF, but not G-AAF. Like an unmodified guanine, G-AF adopts a conformation that allows it to form Watson-Crick hydrogen bonds with an opposing cytosine that results in protrusion of the bulky fluorene moiety into the major groove. Although incorporation opposite G-AF is observed, the C:G-AF base pair induces distortions to the polymerase active site that slow translesion synthesis.


Assuntos
Aminas/metabolismo , Adutos de DNA/biossíntese , DNA Polimerase Dirigida por DNA/metabolismo , DNA/biossíntese , Desoxiguanosina/análogos & derivados
20.
Nature ; 431(7005): 217-21, 2004 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-15322558

RESUMO

Aerobic respiration generates reactive oxygen species that can damage guanine residues and lead to the production of 8-oxoguanine (8oxoG), the major mutagenic oxidative lesion in the genome. Oxidative damage is implicated in ageing and cancer, and its prevalence presents a constant challenge to DNA polymerases that ensure accurate transmission of genomic information. When these polymerases encounter 8oxoG, they frequently catalyse misincorporation of adenine in preference to accurate incorporation of cytosine. This results in the propagation of G to T transversions, which are commonly observed somatic mutations associated with human cancers. Here, we present sequential snapshots of a high-fidelity DNA polymerase during both accurate and mutagenic replication of 8oxoG. Comparison of these crystal structures reveals that 8oxoG induces an inversion of the mismatch recognition mechanisms that normally proofread DNA, such that the 8oxoG.adenine mismatch mimics a cognate base pair whereas the 8oxoG.cytosine base pair behaves as a mismatch. These studies reveal a fundamental mechanism of error-prone replication and show how 8oxoG, and DNA lesions in general, can form mismatches that evade polymerase error-detection mechanisms, potentially leading to the stable incorporation of lethal mutations.


Assuntos
Dano ao DNA , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , DNA/metabolismo , Guanosina/análogos & derivados , Mutagênese , Estresse Oxidativo , Pareamento de Bases/efeitos dos fármacos , Sequência de Bases , Catálise , Cristalografia por Raios X , DNA/biossíntese , DNA/química , DNA/genética , Dano ao DNA/efeitos dos fármacos , Guanosina/metabolismo , Guanosina/farmacologia , Cinética , Modelos Moleculares , Oxirredução , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA