Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Biochemistry ; 57(50): 6827-6837, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30525509

RESUMO

Members of the orthosomycin family of natural products are decorated polysaccharides with potent antibiotic activity and complex biosynthetic pathways. The defining feature of the orthosomycins is an orthoester linkage between carbohydrate moieties that is necessary for antibiotic activity and is likely formed by a family of conserved oxygenases. Everninomicins are octasaccharide orthosomycins produced by Micromonospora carbonacea that have two orthoester linkages and a methylenedioxy bridge, three features whose formation logically requires oxidative chemistry. Correspondingly, the evd gene cluster encoding everninomicin D encodes two monofunctional nonheme iron, α-ketoglutarate-dependent oxygenases and one bifunctional enzyme with an N-terminal methyltransferase domain and a C-terminal oxygenase domain. To investigate whether the activities of these domains are linked in the bifunctional enzyme EvdMO1, we determined the structure of the N-terminal methyltransferase domain to 1.1 Å and that of the full-length protein to 3.35 Å resolution. Both domains of EvdMO1 adopt the canonical folds of their respective superfamilies and are connected by a short linker. Each domain's active site is oriented such that it faces away from the other domain, and there is no evidence of a channel connecting the two. Our results support EvdMO1 working as a bifunctional enzyme with independent catalytic activities.


Assuntos
Aminoglicosídeos/biossíntese , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Metiltransferases/química , Metiltransferases/metabolismo , Micromonospora/enzimologia , Oxigenases/química , Oxigenases/metabolismo , Sequência de Aminoácidos , Aminoglicosídeos/química , Proteínas de Bactérias/genética , Vias Biossintéticas , Domínio Catalítico , Sequência Conservada , Cristalografia por Raios X , Fusão Gênica , Genes Bacterianos , Metiltransferases/genética , Micromonospora/genética , Modelos Moleculares , Oxigenases/genética , Domínios e Motivos de Interação entre Proteínas , Homologia de Sequência de Aminoácidos
2.
RNA ; 22(8): 1250-60, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27277658

RESUMO

Lysine-specific histone demethylase 1 (LSD1) is an essential epigenetic regulator in metazoans and requires the co-repressor element-1 silencing transcription factor (CoREST) to efficiently catalyze the removal of mono- and dimethyl functional groups from histone 3 at lysine positions 4 and 9 (H3K4/9). LSD1 interacts with over 60 regulatory proteins and also associates with lncRNAs (TERRA, HOTAIR), suggesting a regulatory role for RNA in LSD1 function. We report that a stacked, intramolecular G-quadruplex (GQ) forming TERRA RNA (GG[UUAGGG]8UUA) binds tightly to the functional LSD1-CoREST complex (Kd ≈ 96 nM), in contrast to a single GQ RNA unit ([UUAGGG]4U), a GQ DNA ([TTAGGG]4T), or an unstructured single-stranded RNA. Stabilization of a parallel-stranded GQ RNA structure by monovalent potassium ions (K(+)) is required for high affinity binding to the LSD1-CoREST complex. These data indicate that LSD1 can distinguish between RNA and DNA as well as structured versus unstructured nucleotide motifs. Further, cross-linking mass spectrometry identified the primary location of GQ RNA binding within the SWIRM/amine oxidase domain (AOD) of LSD1. An ssRNA binding region adjacent to this GQ binding site was also identified via X-ray crystallography. This RNA binding interface is consistent with kinetic assays, demonstrating that a GQ-forming RNA can serve as a noncompetitive inhibitor of LSD1-catalyzed demethylation. The identification of a GQ RNA binding site coupled with kinetic data suggests that structured RNAs can function as regulatory molecules in LSD1-mediated mechanisms.


Assuntos
Quadruplex G , Histona Desmetilases/metabolismo , Lisina/metabolismo , RNA/metabolismo
3.
J Biol Chem ; 291(14): 7230-40, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-26833566

RESUMO

Streptococcus sanguinisis a leading cause of infective endocarditis, a life-threatening infection of the cardiovascular system. An important interaction in the pathogenesis of infective endocarditis is attachment of the organisms to host platelets.S. sanguinisexpresses a serine-rich repeat adhesin, SrpA, similar in sequence to platelet-binding adhesins associated with increased virulence in this disease. In this study, we determined the first crystal structure of the putative binding region of SrpA (SrpABR) both unliganded and in complex with a synthetic disaccharide ligand at 1.8 and 2.0 Å resolution, respectively. We identified a conserved Thr-Arg motif that orients the sialic acid moiety and is required for binding to platelet monolayers. Furthermore, we propose that sequence insertions in closely related family members contribute to the modulation of structural and functional properties, including the quaternary structure, the tertiary structure, and the ligand-binding site.


Assuntos
Adesinas Bacterianas/química , Ácido N-Acetilneuramínico/química , Streptococcus/química , Fatores de Virulência/química , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Sítios de Ligação , Plaquetas/metabolismo , Plaquetas/microbiologia , Plaquetas/patologia , Endocardite/metabolismo , Endocardite/microbiologia , Endocardite/patologia , Humanos , Ácido N-Acetilneuramínico/genética , Ácido N-Acetilneuramínico/metabolismo , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/patologia , Streptococcus/genética , Streptococcus/metabolismo , Streptococcus/patogenicidade , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
4.
Biochemistry ; 55(42): 5927-5937, 2016 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-27685666

RESUMO

Streptococcus sanguinis is a leading cause of bacterial infective endocarditis, a life-threatening infection of heart valves. S. sanguinis binds to human platelets with high avidity, and this adherence is likely to enhance virulence. Previous studies suggest that a serine-rich repeat adhesin termed SrpA mediates the binding of S. sanguinis to human platelets via its interaction with sialoglycans on the receptor GPIbα. However, in vitro binding assays with SrpA and defined sialoglycans failed to identify specific high-affinity ligands. To improve our understanding of the interaction between SrpA and human platelets, we determined cocrystal structures of the SrpA sialoglycan binding region (SrpABR) with five low-affinity ligands: three sialylated trisaccharides (sialyl-T antigen, 3'-sialyllactose, and 3'-sialyl-N-acetyllactosamine), a sialylated tetrasaccharide (sialyl-LewisX), and a sialyl galactose disaccharide component common to these sialoglyans. We then combined structural analysis with mutagenesis to further determine whether our observed interactions between SrpABR and glycans are important for binding to platelets and to better map the binding site for the physiological receptor. We found that the sialoglycan binding site of SrpABR is significantly larger than the sialoglycans cocrystallized in this study, which suggests that binding of SrpA to platelets either is multivalent or occurs via a larger, disialylated glycan.

5.
Biochem Biophys Res Commun ; 449(4): 392-8, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24858690

RESUMO

Dimethylglycine dehydrogenase (DMGDH) is a mammalian mitochondrial enzyme which plays an important role in the utilization of methyl groups derived from choline. DMGDH is a flavin containing enzyme which catalyzes the oxidative demethylation of dimethylglycine in vitro with the formation of sarcosine (N-methylglycine), hydrogen peroxide and formaldehyde. DMGDH binds tetrahydrofolate (THF) in vivo, which serves as an acceptor of formaldehyde and in the cell the product of the reaction is 5,10-methylenetetrahydrofolate instead of formaldehyde. To gain insight into the mechanism of the reaction we solved the crystal structures of the recombinant mature and precursor forms of rat DMGDH and DMGDH-THF complexes. Both forms of DMGDH reveal similar kinetic parameters and have the same tertiary structure fold with two domains formed by N- and C-terminal halves of the protein. The active center is located in the N-terminal domain while the THF binding site is located in the C-terminal domain about 40Å from the isoalloxazine ring of FAD. The folate binding site is connected with the enzyme active center via an intramolecular channel. This suggests the possible transfer of the intermediate imine of dimethylglycine from the active center to the bound THF where they could react producing a 5,10-methylenetetrahydrofolate. Based on the homology of the rat and human DMGDH the structural basis for the mechanism of inactivation of the human DMGDH by naturally occurring His109Arg mutation is proposed.


Assuntos
Dimetilglicina Desidrogenase/química , Proteínas Mitocondriais/química , Tetra-Hidrofolatos/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Domínio Catalítico , Cristalização , Cristalografia por Raios X , Dimetilglicina Desidrogenase/metabolismo , Humanos , Cinética , Proteínas Mitocondriais/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Ratos , Sarcosina/análogos & derivados , Tetra-Hidrofolatos/metabolismo
6.
Biochim Biophys Acta ; 1824(2): 286-91, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22037183

RESUMO

Glycine N-methyltransferase (GNMT) is a key regulatory enzyme in methyl group metabolism. In mammalian liver it reduces S-adenosylmethionine levels by using it to methylate glycine, producing N-methylglycine (sarcosine) and S-adenosylhomocysteine. GNMT is inhibited by binding two molecules of 5-methyltetrahydrofolate (mono- or polyglutamate forms) per tetramer of the active enzyme. Inhibition is sensitive to the status of the N-terminal valine of GNMT and to polyglutamation of the folate inhibitor. It is inhibited by pentaglutamate form more efficiently compared to monoglutamate form. The native rat liver GNMT contains an acetylated N-terminal valine and is inhibited much more efficiently compared to the recombinant protein expressed in E. coli where the N-terminus is not acetylated. In this work we used a protein crystallography approach to evaluate the structural basis for these differences. We show that in the folate-GNMT complexes with the native enzyme, two folate molecules establish three and four hydrogen bonds with the protein. In the folate-recombinant GNMT complex only one hydrogen bond is established. This difference results in more effective inhibition by folate of the native liver GNMT activity compared to the recombinant enzyme.


Assuntos
Glicina N-Metiltransferase/metabolismo , Fígado/enzimologia , Modelos Moleculares , Proteínas Recombinantes/metabolismo , Animais , Domínio Catalítico , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Glicina N-Metiltransferase/antagonistas & inibidores , Glicina N-Metiltransferase/química , Glicina N-Metiltransferase/isolamento & purificação , Ligação de Hidrogênio , Ligação Proteica , Ratos , Proteínas Recombinantes/química , Tetra-Hidrofolatos/química , Tetra-Hidrofolatos/metabolismo , Valina/metabolismo
7.
Biochemistry ; 50(21): 4750-6, 2011 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-21510664

RESUMO

Methylation of lysine residues in histones has been known to serve a regulatory role in gene expression. Although enzymatic removal of the methyl groups was discovered as early as 1973, the enzymes responsible for their removal were isolated and their mechanism of action was described only recently. The first enzyme to show such activity was LSD1, a flavin-containing enzyme that removes the methyl groups from lysines 4 and 9 of histone 3 with the generation of formaldehyde from the methyl group. This reaction is similar to the previously described demethylation reactions conducted by the enzymes dimethylglycine dehydrogenase and sarcosine dehydrogenase, in which protein-bound tetrahydrofolate serves as an accepter of the formaldehyde that is generated. We now show that nuclear extracts of HeLa cells contain LSD1 that is associated with folate. Using the method of back-scattering interferometry, we have measured the binding of various forms of folate to both full-length LSD1 and a truncated form of LSD1 in free solution. The 6R,S form of the natural pentaglutamate form of tetrahydrofolate bound with the highest affinity (K(d) = 2.8 µM) to full-length LSD1. The fact that folate participates in the enzymatic demethylation of histones provides an opportunity for this micronutrient to play a role in the epigenetic control of gene expression.


Assuntos
Ácido Fólico/metabolismo , Histona Desmetilases/metabolismo , Cromatografia em Gel , Células HeLa , Humanos
8.
Artigo em Inglês | MEDLINE | ID: mdl-20823515

RESUMO

Crystal structures of a binary Mg2+-form Dpo4-DNA complex with 1,N2-etheno-dG in the template strand as well as of ternary Mg2+-form Dpo4-DNA-dCTP/dGTP complexes with 8-oxoG in the template strand have been determined. Comparison of their conformations and active-site geometries with those of the corresponding Ca2+-form complexes revealed that the DNA and polymerase undergo subtle changes as a result of the catalytically more active Mg2+ occupying both the A and B sites.


Assuntos
Cálcio/química , Domínio Catalítico , DNA Polimerase beta/química , Magnésio/química , Sulfolobus solfataricus/enzimologia , Biocatálise , Cálcio/metabolismo , Cátions Bivalentes/química , Cátions Bivalentes/metabolismo , Cristalografia por Raios X , Magnésio/metabolismo , Modelos Moleculares , Ligação Proteica , Homologia Estrutural de Proteína
9.
Biochim Biophys Acta ; 1784(9): 1342-6, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18501206

RESUMO

Native liver glycine N-methyltransferase (GNMT) is N-acetylated while the recombinant enzyme is not. We show here that acetylation of the N-terminal valine affects several kinetic parameters of the enzyme. Glycine N-methyltransferase is a regulatory enzyme mediating the availability of methyl groups by virtue of being inhibited by folate. N-acetylation does not affect the overall structure of the protein and does not affect basal enzyme activity of GNMT. Binding of both the mono- and pentaglutamate forms of 5-methyltetrahydrofolate is the same for the acetylated and non-acetylated forms of the enzyme, however the pentaglutamate form is bound more tightly than the monoglutamate form in both cases. Although binding of the folates is similar for the acetylated and non-acetylated forms of the enzyme, inhibition of enzyme activity differs significantly. The native, N-acetylated form of the enzyme shows 50% inhibition at 1.3 microM concentration of the pentaglutamate while the recombinant non-acetylated form shows 50% inhibition at 590 microM. In addition, the binding of folate results in cooperativity of the substrate S-adenosylmethionine (AdoMet), with a Hill coefficient of 1.5 for 5-methyltetrahydrofolate pentaglutamate.


Assuntos
Ácido Fólico/farmacologia , Glicina N-Metiltransferase/antagonistas & inibidores , Glicina N-Metiltransferase/química , Acetilação , Regulação Alostérica , Animais , Inibidores Enzimáticos/farmacologia , Glicina N-Metiltransferase/genética , Glicina N-Metiltransferase/metabolismo , Técnicas In Vitro , Cinética , Fígado/enzimologia , Modelos Moleculares , Conformação Proteica , Ratos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Valina/química
10.
Artigo em Inglês | MEDLINE | ID: mdl-17277445

RESUMO

RNase H binds RNA-DNA hybrid and double-stranded RNA (dsRNA) duplexes with similar affinity, but only cleaves the RNA in the former. To potentially gain insight into the conformational origins of substrate recognition by the enzyme from Escherichia coli, cocrystallization experiments were carried out with RNase HI-dsRNA (enzyme-inhibitor) complexes. Crystals were obtained of two complexes containing 9-mer and 10-mer RNA duplexes that diffracted X-rays to 3.5 and 4 A resolution, respectively.


Assuntos
Escherichia coli/química , RNA de Cadeia Dupla/química , Ribonuclease H/química , Cristalografia por Raios X , Escherichia coli/enzimologia , Escherichia coli/genética , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , Ribonuclease H/genética , Ribonuclease H/metabolismo
11.
Med Hypotheses ; 88: 63-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26880641

RESUMO

Formaldehyde is extremely toxic reacting with proteins to crosslinks peptide chains. Formaldehyde is a metabolic product in many enzymatic reactions and the question of how these enzymes are protected from the formaldehyde that is generated has largely remained unanswered. Early experiments from our laboratory showed that two liver mitochondrial enzymes, dimethylglycine dehydrogenase (DMGDH) and sarcosine dehydrogenase (SDH) catalyze oxidative demethylation reactions (sarcosine is a common name for monomethylglycine). The enzymatic products of these enzymes were the demethylated substrates and formaldehyde, produced from the removed methyl group. Both DMGDH and SDH contain FAD and both have tightly bound tetrahydrofolate (THF), a folate coenzyme. THF binds reversibly with formaldehyde to form 5,10-methylene-THF. At that time we showed that purified DMGDH, with tightly bound THF, reacted with formaldehyde generated during the reaction to form 5,10-methylene-THF. This effectively scavenged the formaldehyde to protect the enzyme. Recently, post-translational modifications on histone tails have been shown to be responsible for epigenetic regulation of gene expression. One of these modifications is methylation of lysine residues. The first enzyme discovered to accomplish demethylation of these modified histones was histone lysine demethylase (LSD1). LSD1 specifically removes methyl groups from di- and mono-methylated lysines at position 4 of histone 3. This enzyme contained tightly bound FAD and the products of the reaction were the demethylated lysine residue and formaldehyde. The mechanism of LSD1 demethylation is analogous to the mechanism previously postulated for DMGDH, i.e. oxidation of the N-methyl bond to the methylene imine followed by hydrolysis to generate formaldehyde. This suggested that THF might also be involved in the LSD1 reaction to scavenge the formaldehyde produced. Our hypotheses are that THF is bound to native LSD1 by analogy to DMGDH and SDH and that the bound THF serves to protect the FAD class of histone demethylases from the destructive effects of formaldehyde generation by formation of 5,10-methylene-THF. We present pilot data showing that decreased folate in livers as a result of dietary folate deficiency is associated with increased levels of methylated lysine 4 of histone 3. This can be a result of decreased LSD1 activity resulting from the decreased folate available to scavenge the formaldehyde produced at the active site caused by the folate deficiency. Because LSD1 can regulate gene expression this suggests that folate may play a more important role than simply serving as a carrier of one-carbon units and be a factor in other diseases associated with low folate.


Assuntos
Metilação de DNA , Deficiência de Ácido Fólico/imunologia , Histona Desmetilases/química , Histonas/química , Domínio Catalítico , Epigênese Genética , Escherichia coli/metabolismo , Ácido Fólico/química , Humanos , Lisina/química , Espectrometria de Massas , Modelos Teóricos , Projetos Piloto , Processamento de Proteína Pós-Traducional , Sarcosina Desidrogenase/química , Tetra-Hidrofolatos/química
12.
Am J Cancer Res ; 5(1): 458-65, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25628954

RESUMO

Methionine metabolism is an important component of one-carbon metabolism. S-adenosylmethionine (SAM), the methyl donor for nearly all methylation reactions, is irreversibly converted to S-adenosylhomocysteine (SAH), an inhibitor of methyltransferases, some of which are key enzymes for methylation. Changes in DNA methylation are common in colorectal cancers. We evaluated plasma SAM and SAH with colorectal adenoma risk in a matched case-control study conducted among individuals undergoing routine colonoscopy. 216 cases were individually matched to polyp-free controls in a 1:1 ratio on age (± 5 years), sex, race (white/non-white), study site (academic medical center/VA hospital) and date of sample collection (± 60 days). Sex-specific quantiles were evaluated based on the control distribution due to vastly different metabolite levels by sex. Conditional logistic regression models were used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). Among males, both higher SAM (OR = 0.38, 95% CI: 0.18-0.77, p for trend = 0.007) and higher SAH (OR = 0.45, 95% CI: 0.22-0.91, p for trend = 0.02) were associated with statistically significantly decreased risks of colorectal adenoma in comparison to lowest plasma SAM or SAH tertile. Conversely, among females, both higher SAM and higher SAH were associated with increased risk of colorectal adenoma, which was statistically significant for SAH (OR = 5.18, 95% CI: 1.09-24.62, p for trend = 0.04). The difference in these associations between men and women was statistically significant (p < 0.05). The ratio of SAM/SAH was not associated with colorectal adenoma risk among males or females. These findings suggest SAM and SAH may be involved in the development of colorectal adenoma and the association may be modified by sex.

13.
Protein Sci ; 23(7): 993-8, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24715612

RESUMO

An important epigenetic modification is the methylation/demethylation of histone lysine residues. The first histone demethylase to be discovered was a lysine-specific demethylase 1, LSD1, a flavin containing enzyme which carries out the demethylation of di- and monomethyllysine 4 in histone H3. The removed methyl groups are oxidized to formaldehyde. This reaction is similar to those performed by dimethylglycine dehydrogenase and sarcosine dehydrogenase, in which protein-bound tetrahydrofolate (THF) was proposed to serve as an acceptor of the generated formaldehyde. We showed earlier that LSD1 binds THF with high affinity which suggests its possible participation in the histone demethylation reaction. In the cell, LSD1 interacts with co-repressor for repressor element 1 silencing transcription factor (CoREST). In order to elucidate the role of folate in the demethylating reaction we solved the crystal structure of the LSD1-CoREST-THF complex. In the complex, the folate-binding site is located in the active center in close proximity to flavin adenine dinucleotide. This position of the folate suggests that the bound THF accepts the formaldehyde generated in the course of histone demethylation to form 5,10-methylene-THF. We also show the formation of 5,10-methylene-THF during the course of the enzymatic reaction in the presence of THF by mass spectrometry. Production of this form of folate could act to prevent accumulation of potentially toxic formaldehyde in the cell. These studies suggest that folate may play a role in the epigenetic control of gene expression in addition to its traditional role in the transfer of one-carbon units in metabolism.


Assuntos
Proteínas Correpressoras/química , Histona Desmetilases/química , Lisina/metabolismo , Tetra-Hidrofolatos/química , Sítios de Ligação , Proteínas Correpressoras/metabolismo , Cristalografia por Raios X , Flavina-Adenina Dinucleotídeo/química , Histona Desmetilases/metabolismo , Humanos , Espectrometria de Massas , Modelos Moleculares , Conformação Proteica , Especificidade por Substrato , Tetra-Hidrofolatos/metabolismo
14.
PLoS One ; 6(8): e22486, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21853037

RESUMO

BACKGROUND: Clinical decision for primary treatment for prostate cancer is dictated by variables with insufficient specificity. Early detection of prostate cancer likely to develop rapid recurrence could support neo-adjuvant therapeutics and adjuvant options prior to frank biochemical recurrence. This study compared markers in serum and urine of patients with rapidly recurrent prostate cancer to recurrence-free patients after radical prostatectomy. Based on previous identification of urinary sarcosine as a metastatic marker, we tested whether methionine metabolites in urine and serum could serve as pre-surgical markers for aggressive disease. METHODOLOGY/PRINCIPAL FINDINGS: Urine and serum samples (n = 54 and 58, respectively), collected at the time of prostatectomy were divided into subjects who developed biochemical recurrence within 2 years and those who remained recurrence-free after 5 years. Multiple methionine metabolites were measured in urine and serum by GC-MS. The role of serum metabolites and clinical variables (biopsy Gleason grade, clinical stage, serum prostate specific antigen [PSA]) on biochemical recurrence prediction were evaluated. Urinary sarcosine and cysteine levels were significantly higher (p = 0.03 and p = 0.007 respectively) in the recurrent group. However, in serum, concentrations of homocysteine (p = 0.003), cystathionine (p = 0.007) and cysteine (p<0.001) were more abundant in the recurrent population. The inclusion of serum cysteine to a model with PSA and biopsy Gleason grade improved prediction over the clinical variables alone (p<0.001). CONCLUSIONS: Higher serum homocysteine, cystathionine, and cysteine concentrations independently predicted risk of early biochemical recurrence and aggressiveness of disease in a nested case control study. The methionine metabolites further supplemented known clinical variables to provide superior sensitivity and specificity in multivariable prediction models for rapid biochemical recurrence following prostatectomy.


Assuntos
Progressão da Doença , Metionina/sangue , Neoplasias da Próstata/sangue , Neoplasias da Próstata/patologia , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/urina , Humanos , Estimativa de Kaplan-Meier , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Modelos de Riscos Proporcionais , Neoplasias da Próstata/cirurgia , Neoplasias da Próstata/urina , Curva ROC , Recidiva , Fatores de Risco
15.
J Biol Chem ; 282(6): 4069-75, 2007 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-17158459

RESUMO

Glycine N-methyltransferase (GNMT) is a key regulatory enzyme in methyl group metabolism. It is abundant in the liver, where it uses excess S-adenosylmethionine (AdoMet) to methylate glycine to N-methylglycine (sarcosine) and produces S-adenosylhomocysteine (AdoHcy), thereby controlling the methylating potential of the cell. GNMT also links utilization of preformed methyl groups, in the form of methionine, to their de novo synthesis, because it is inhibited by a specific form of folate, 5-methyltetrahydrofolate. Although the structure of the enzyme has been elucidated by x-ray crystallography of the apoenzyme and in the presence of the substrate, the location of the folate inhibitor in the tetrameric structure has not been identified. We report here for the first time the crystal structure of rat GNMT complexed with 5-methyltetrahydrofolate. In the GNMT-folate complex, two folate binding sites were located in the intersubunit areas of the tetramer. Each folate binding site is formed primarily by two 1-7 N-terminal regions of one pair of subunits and two 205-218 regions of the other pair of subunits. Both the pteridine and p-aminobenzoyl rings are located in the hydrophobic cavities formed by Tyr5, Leu207, and Met215 residues of all subunits. Binding experiments in solution also confirm that one GNMT tetramer binds two folate molecules. For the enzymatic reaction to take place, the N-terminal fragments of GNMT must have a significant degree of conformational freedom to provide access to the active sites. The presence of the folate in this position provides a mechanism for its inhibition.


Assuntos
Proteínas de Transporte/metabolismo , Ácido Fólico/metabolismo , Glicina N-Metiltransferase/metabolismo , Fígado/enzimologia , Subunidades Proteicas/metabolismo , Receptores de Superfície Celular/metabolismo , Tetra-Hidrofolatos/metabolismo , Animais , Sítios de Ligação , Proteínas de Transporte/química , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Receptores de Folato com Âncoras de GPI , Ácido Fólico/química , Glicina N-Metiltransferase/antagonistas & inibidores , Glicina N-Metiltransferase/química , Fígado/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Subunidades Proteicas/química , Ratos , Receptores de Superfície Celular/química , Tetra-Hidrofolatos/química
16.
Biochemistry ; 45(19): 5949-56, 2006 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-16681366

RESUMO

Y-Family DNA polymerase IV (Dpo4) from Sulfolobus solfataricus serves as a model system for eukaryotic translesion polymerases, and three-dimensional structures of its complexes with native and adducted DNA have been analyzed in considerable detail. Dpo4 lacks a proofreading exonuclease activity common in replicative polymerases but uses pyrophosphorolysis to reduce the likelihood of incorporation of an incorrect base. Mg(2+) is a cofactor for both the polymerase and pyrophosphorolysis activities. Despite the fact that all crystal structures of Dpo4 have been obtained in the presence of Ca(2+), the consequences of replacing Mg(2+) with Ca(2+) for Dpo4 activity have not been investigated to date. We show here that Ca(2+) (but not Ba(2+), Co(2+), Cu(2+), Ni(2+), or Zn(2+)) is a cofactor for Dpo4-catalyzed polymerization with both native and 8-oxoG-containing DNA templates. Both dNTP and ddNTP are substrates of the polymerase in the presence of either Mg(2+) or Ca(2+). Conversely, no pyrophosphorolysis occurs in the presence of Ca(2+), although the positions of the two catalytic metal ions at the active site appear to be very similar in mixed Mg(2+)/Ca(2+)- and Ca(2+)-form Dpo4 crystals.


Assuntos
Biopolímeros/metabolismo , Cálcio/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Difosfatos/metabolismo , Sulfolobus solfataricus/metabolismo , Sequência de Bases , Cátions Bivalentes/metabolismo , Primers do DNA , Modelos Moleculares
17.
J Biol Chem ; 281(4): 2358-72, 2006 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-16306039

RESUMO

DNA polymerases insert dATP opposite the oxidative damage product 7,8-dihydro-8-oxodeoxyguanosine (8-oxoG) instead of dCTP, to the extent of >90% with some polymerases. Steady-state kinetics with the Y-family Sulfolobus solfataricus DNA polymerase IV (Dpo4) showed 90-fold higher incorporation efficiency of dCTP > dATP opposite 8-oxoG and 4-fold higher efficiency of extension beyond an 8-oxoG:C pair than an 8-oxoG:A pair. The catalytic efficiency for these events (with dCTP or C) was similar for G and 8-oxoG templates. Mass spectral analysis of extended DNA primers showed >/=95% incorporation of dCTP > dATP opposite 8-oxoG. Pre-steady-state kinetics showed faster rates of dCTP incorporation opposite 8-oxoG than G. The measured K(d)(,dCTP) was 15-fold lower for an oligonucleotide containing 8-oxoG than with G. Extension beyond an 8-oxoG:C pair was similar to G:C and faster than for an 8-oxoG:A pair, in contrast to other polymerases. The E(a) for dCTP insertion opposite 8-oxoG was lower than for opposite G. Crystal structures of Dpo4 complexes with oligonucleotides were solved with C, A, and G nucleoside triphosphates placed opposite 8-oxoG. With ddCTP, dCTP, and dATP the phosphodiester bonds were formed even in the presence of Ca(2+). The 8-oxoG:C pair showed classic Watson-Crick geometry; the 8-oxoG:A pair was in the syn:anti configuration, with the A hybridized in a Hoogsteen pair with 8-oxoG. With dGTP placed opposite 8-oxoG, pairing was not to the 8-oxoG but to the 5' C (and in classic Watson-Crick geometry), consistent with the low frequency of this frameshift event observed in the catalytic assays.


Assuntos
DNA Polimerase Dirigida por DNA/química , DNA Polimerase Dirigida por DNA/metabolismo , DNA Polimerase Dirigida por DNA/fisiologia , Nucleotídeos de Desoxicitosina/química , Desoxiguanosina/análogos & derivados , Sulfolobus solfataricus/enzimologia , 8-Hidroxi-2'-Desoxiguanosina , Sequência de Bases , Cromatografia Líquida , DNA/química , Primers do DNA/química , Desoxiguanosina/química , Relação Dose-Resposta a Droga , Escherichia coli/metabolismo , Mutação da Fase de Leitura , Cinética , Espectrometria de Massas , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Fatores de Tempo , Difração de Raios X
18.
J Biol Chem ; 280(33): 29750-64, 2005 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-15965231

RESUMO

1,N(2)-Etheno(epsilon)guanine is a mutagenic DNA lesion derived from lipid oxidation products and also from some chemical carcinogens. Gel electrophoretic analysis of the products of primer extension by Sulfolobus solfataricus P2 DNA polymerase IV (Dpo4) indicated preferential incorporation of A opposite 3'-(1,N(2)-epsilon-G)TACT-5', among the four dNTPs tested individually. With the template 3'-(1,N(2)-epsilon-G)CACT-5', both G and A were incorporated. When primer extension was done in the presence of a mixture of all four dNTPs, high pressure liquid chromatography-mass spectrometry analysis of the products indicated that (opposite 3'-(1,N(2)-epsilon-G)CACT-5') the major product was 5'-GTGA-3' and the minor product was 5'-AGTGA-3'. With the template 3'-(1,N(2)-epsilon-G)TACT-5', the following four products were identified by high pressure liquid chromatography-mass spectrometry: 5'-AATGA-3', 5'-ATTGA-3', 5'-ATGA-3', and 5'-TGA-3'. An x-ray crystal structure of Dpo4 was solved (2.1 A) with a primer-template and A placed in the primer to be opposite the 1,N(2)-epsilon-G in the template 3'-(1,N(2)-epsilon-G)TACT 5'. The added A in the primer was paired across the template T with classic Watson-Crick geometry. Similar structures were observed in a ternary Dpo4-DNA-dATP complex and a ternary Dpo4-DNA-ddATP complex, with d(d)ATP opposite the template T. A similar structure was observed with a ddGTP adjacent to the primer and opposite the C next to 1,N(2)-epsilon-G in 3'-(1,N(2)-epsilon-G)CACT-5'. We concluded that Dpo4 uses several mechanisms, including A incorporation opposite 1,N(2)-epsilon-G and also a variation of dNTP-stabilized misalignment, to generate both base pair and frameshift mutations.


Assuntos
Adutos de DNA/química , DNA Polimerase beta/química , Sulfolobus solfataricus/enzimologia , Sítios de Ligação , Cristalografia por Raios X , DNA Polimerase beta/fisiologia , Primers do DNA/metabolismo , Mutação da Fase de Leitura , Guanina/metabolismo , Espectrometria de Massas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA