Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 255
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Med Microbiol ; 313(2): 151577, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36841056

RESUMO

Staphylococcus aureus (S. aureus) is one of the critical clinical pathogens which can cause multiple diseases ranging from skin infections to fatal sepsis. S. aureus is generally considered to be an extracellular pathogen. However, more and more evidence has shown that S. aureus can survive inside various cells. Folate plays an essential role in multiple life activities, including the conversion of serine and glycine, the remethylation of homocysteine to methionine, and the de novo synthesis of purine /dTMP, et al. More and more studies reported that S. aureus intracellular infection requires the involvement of folate metabolism. This review focused on the mechanisms of folate metabolism and related substances affecting S. aureus infection. Loss of tetrahydrofolic acid (THF)-dependent dTMP directly inhibits the nucleotide synthesis pathway of the S. aureus due to pabA deficiency. Besides, trimethoprim-sulfamethoxazole (TMP/SMX), a potent antibiotic that treats S. aureus infections, interferes in the process of the folate mechanism and leads to the production of thymidine-dependent small-colony variants (TD-SCVs). In addition, S. aureus is resistant to lysostaphin in the presence of serine hydroxymethyltransferase (SHMT). We provide new insights for understanding the molecular pathogenesis of S. aureus infection.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Staphylococcus aureus/metabolismo , Timidina Monofosfato/metabolismo , Timidina Monofosfato/uso terapêutico , Infecções Estafilocócicas/tratamento farmacológico , Combinação Trimetoprima e Sulfametoxazol/farmacologia , Combinação Trimetoprima e Sulfametoxazol/uso terapêutico , Ácido Fólico/metabolismo , Ácido Fólico/uso terapêutico
2.
Pain Pract ; 23(6): 639-646, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37067033

RESUMO

BACKGROUND: Spinal cord stimulation (SCS) has been proven to be an effective treatment for patients suffering from intractable chronic neuropathic pain. Recent advances in the field include the utilization of programs that multiplex various signals to target different neural structures in the dorsal spinal cord associated with the painful area. Preclinical studies have been fundamental in understanding the mechanism by which this differential target multiplexed programming (DTMP) SCS approach works. Transcriptomic- and proteomic-based studies demonstrated that DTMP can modulate expression levels of genes and proteins involved in pain-related processes that have been affected by a neuropathic pain model. This work studied the effect of the intensity of DTMP signals on mechanical hypersensitivity and cell-specific transcriptomes. METHODS: The spared nerve injury model (SNI) of neuropathic pain was induced in 20 animals which were 1:1 randomized into two SCS groups in which the intensity of the DTMP was adjusted to either 70% or 40% of the motor threshold (MT). SCS was applied continuously for 48 h via a quadripolar lead implanted in the dorsal epidural space of animals. Controls, which included a group of implanted SNI animals that received no SCS and a group of animals naive to the SNI, were assessed in parallel to the SCS groups. Mechanical hypersensitivity was assessed before SNI, before SCS, and at 48 h of SCS. At the end of SCS, the stimulated segment of the dorsal spinal cord was dissected and subjected to RNA sequencing to quantify expression levels in all experimental groups. Differential effects were assessed via fold-change comparisons of SCS and naive groups versus the no-SCS group for transcriptomes specific to neurons and glial cells. Standard statistical analyses were employed to assess significance of the comparisons (p < 0.05). RESULTS: SCS treatments provided significant improvement in mechanical sensitivity relative to no SCS treatment. However, the change in the intensity did not provide a significant difference in the improvement of mechanical sensitivity. DTMP regulated expression levels back toward those found in the naive group in the cell-specific transcriptomes analyzed. There were no significant differences related to the intensity of the stimulation in terms of the percentage of genes in each transcriptome in which expression levels were reversed toward the naive state. CONCLUSIONS: DTMP when applied at either 40% MT or 70% MT provided similar reduction of pain-like behavior in rats and similar effects in neuron- and glia-specific transcriptomes.


Assuntos
Neuralgia , Estimulação da Medula Espinal , Ratos , Animais , Limiar da Dor/fisiologia , Medição da Dor , Proteômica , Timidina Monofosfato/metabolismo , Modelos Animais de Doenças , Neuralgia/terapia , Neuralgia/metabolismo , Medula Espinal/fisiologia
3.
J Biol Chem ; 293(52): 20285-20294, 2018 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-30385507

RESUMO

Mitochondrial inner membrane protein MPV17 is a protein of unknown function that is associated with mitochondrial DNA (mtDNA)-depletion syndrome (MDS). MPV17 loss-of-function has been reported to result in tissue-specific nucleotide pool imbalances, which can occur in states of perturbed folate-mediated one-carbon metabolism (FOCM), but MPV17 has not been directly linked to FOCM. FOCM is a metabolic network that provides one-carbon units for the de novo synthesis of purine and thymidylate nucleotides (e.g. dTMP) for both nuclear DNA (nuDNA) and mtDNA replication. In this study, we investigated the impact of reduced MPV17 expression on markers of impaired FOCM in HeLa cells. Depressed MPV17 expression reduced mitochondrial folate levels by 43% and increased uracil levels, a marker of impaired dTMP synthesis, in mtDNA by 3-fold. The capacity of mitochondrial de novo and salvage pathway dTMP biosynthesis was unchanged by the reduced MPV17 expression, but the elevated levels of uracil in mtDNA suggested that other sources of mitochondrial dTMP are compromised in MPV17-deficient cells. These results indicate that MPV17 provides a third dTMP source, potentially by serving as a transporter that transfers dTMP from the cytosol to mitochondria to sustain mtDNA synthesis. We propose that MPV17 loss-of-function and related hepatocerebral MDS are linked to impaired FOCM in mitochondria by providing insufficient access to cytosolic dTMP pools and by severely reducing mitochondrial folate pools.


Assuntos
DNA Mitocondrial/biossíntese , Regulação da Expressão Gênica , Proteínas de Membrana/biossíntese , Doenças Mitocondriais/metabolismo , Proteínas Mitocondriais/biossíntese , Uracila/metabolismo , Transporte Biológico Ativo/genética , DNA Mitocondrial/genética , Ácido Fólico/genética , Ácido Fólico/metabolismo , Células HeLa , Humanos , Proteínas de Membrana/genética , Doenças Mitocondriais/genética , Doenças Mitocondriais/patologia , Proteínas Mitocondriais/genética , Timidina Monofosfato/genética , Timidina Monofosfato/metabolismo
4.
Annu Rev Nutr ; 38: 219-243, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30130467

RESUMO

Despite unequivocal evidence that folate deficiency increases risk for human pathologies, and that folic acid intake among women of childbearing age markedly decreases risk for birth defects, definitive evidence for a causal biochemical pathway linking folate to disease and birth defect etiology remains elusive. The de novo and salvage pathways for thymidylate synthesis translocate to the nucleus of mammalian cells during S- and G2/M-phases of the cell cycle and associate with the DNA replication and repair machinery, which limits uracil misincorporation into DNA and genome instability. There is increasing evidence that impairments in nuclear de novo thymidylate synthesis occur in many pathologies resulting from impairments in one-carbon metabolism. Understanding the roles and regulation of nuclear de novo thymidylate synthesis and its relationship to genome stability will increase our understanding of the fundamental mechanisms underlying folate- and vitamin B12-associated pathologies.


Assuntos
Núcleo Celular/metabolismo , Ácido Fólico/metabolismo , Animais , Ciclo Celular , Regulação da Expressão Gênica/fisiologia , Humanos , Timidina Monofosfato/metabolismo
5.
Molecules ; 24(7)2019 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-30935102

RESUMO

Thymidylate synthase (TS) is an enzyme of paramount importance as it provides the only de novo source of deoxy-thymidine monophosphate (dTMP). dTMP, essential for DNA synthesis, is produced by the TS-catalyzed reductive methylation of 2'-deoxyuridine-5'-monophosphate (dUMP) using N5,N10-methylenetetrahydrofolate (mTHF) as a cofactor. TS is ubiquitous and a validated drug target. TS enzymes from different organisms differ in sequence and structure, but are all obligate homodimers. The structural and mechanistic differences between the human and bacterial enzymes are exploitable to obtain selective inhibitors of bacterial TSs that can enrich the currently available therapeutic tools against bacterial infections. Enterococcus faecalis is a pathogen fully dependent on TS for dTMP synthesis. In this study, we present four new crystal structures of Enterococcus faecalis and human TSs in complex with either the substrate dUMP or the inhibitor FdUMP. The results provide new clues about the half-site reactivity of Enterococcus faecalis TS and the mechanisms underlying the conformational changes occurring in the two enzymes. We also identify relevant differences in cofactor and inhibitor binding between Enterococcus faecalis and human TS that can guide the design of selective inhibitors against bacterial TSs.


Assuntos
Enterococcus faecalis/enzimologia , Fluordesoxiuridilato/química , Conformação Proteica , Timidina Monofosfato/química , Timidilato Sintase/química , Sítios de Ligação , Domínio Catalítico , Fluordesoxiuridilato/metabolismo , Humanos , Modelos Moleculares , Ligação Proteica , Multimerização Proteica , Relação Estrutura-Atividade , Especificidade por Substrato , Timidina Monofosfato/metabolismo , Timidilato Sintase/metabolismo
6.
Mol Microbiol ; 102(3): 365-385, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27426054

RESUMO

Thymidine kinase (TK) is a key enzyme in the pyrimidine salvage pathway which catalyzes the transfer of the γ-phosphate of ATP to 2'-deoxythymidine (dThd) forming thymidine monophosphate (dTMP). Unlike other type II TKs, the Trypanosoma brucei enzyme (TbTK) is a tandem protein with two TK homolog domains of which only the C-terminal one is active. In this study, we establish that TbTK is essential for parasite viability and cell cycle progression, independently of extracellular pyrimidine concentrations. We show that expression of TbTK is cell cycle regulated and that depletion of TbTK leads to strongly diminished dTTP pools and DNA damage indicating intracellular dThd to be an essential intermediate metabolite for the synthesis of thymine-derived nucleotides. In addition, we report the X-ray structure of the catalytically active domain of TbTK in complex with dThd and dTMP at resolutions up to 2.2 Å. In spite of the high conservation of the active site residues, the structures reveal a widened active site cavity near the nucleobase moiety compared to the human enzyme. Our findings strongly support TbTK as a crucial enzyme in dTTP homeostasis and identify structural differences within the active site that could be exploited in the process of rational drug design.


Assuntos
Timidina Quinase/metabolismo , Trypanosoma brucei brucei/citologia , Trypanosoma brucei brucei/enzimologia , Pontos de Checagem do Ciclo Celular/fisiologia , Núcleosídeo-Fosfato Quinase/metabolismo , Relação Estrutura-Atividade , Timidina/metabolismo , Timidina Quinase/química , Timidina Monofosfato/metabolismo , Nucleotídeos de Timina/metabolismo , Trypanosoma brucei brucei/metabolismo
7.
Biochemistry ; 54(5): 1287-93, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25581782

RESUMO

The development of cancer-specific probes for imaging by positron emission tomography (PET) is gaining impetus in cancer research and clinical oncology. One of the hallmarks of most cancer cells is incessant DNA replication, which requires the continuous synthesis of nucleotides. Thymidylate synthase (TSase) is unique in this context because it is the only enzyme in humans that is responsible for the de novo biosynthesis of the DNA building block 2'-deoxy-thymidylate (dTMP). TSase catalyzes the reductive methylation of 2'-deoxy-uridylate (dUMP) to dTMP using (R)-N(5),N(10)-methylene-5,6,7,8-tetrahydrofolate (MTHF) as a cofactor. Not surprisingly, several human cancers overexpress TSase, which makes it a common target for chemotherapy (e.g., 5-fluorouracil). We envisioned that [(11)C]-MTHF might be a PET probe that could specifically label cancerous cells. Using stable radiotracer [(14)C]-MTHF, we had initially found increased uptake by breast and colon cancer cell lines. In the current study, we examined the uptake of this radiotracer in human pancreatic cancer cell lines MIAPaCa-2 and PANC-1 and found predominant radiolabeling of cancerous versus normal pancreatic cells. Furthermore, uptake of the radiotracer is dependent on the intracellular level of the folate pool, cell cycle phase, expression of folate receptors on the cell membrane, and cotreatment with the common chemotherapeutic drug methotrexate (MTX, which blocks the biosynthesis of endogenous MTHF). These results point toward [(11)C]-MTHF being used as PET probe with broad specificity and being able to control its signal through MTX co-administration.


Assuntos
Neoplasias Pancreáticas , Tomografia por Emissão de Pósitrons/métodos , Traçadores Radioativos , Timidina Monofosfato/biossíntese , Timidilato Sintase/biossíntese , Isótopos de Carbono , Ácido Fólico/análogos & derivados , Ácido Fólico/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Marcação por Isótopo , Neoplasias Pancreáticas/diagnóstico por imagem , Neoplasias Pancreáticas/metabolismo , Radiografia , Timidina Monofosfato/metabolismo , Uridina Monofosfato/metabolismo
8.
Mol Cell Biochem ; 398(1-2): 223-31, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25245820

RESUMO

The causal metabolic pathway and the underlying mechanism between folate deficiency and neural tube defects (NTDs) remain obscure. Thymidylate (dTMP) is catalyzed by thymidylate synthase (TS) using the folate-derived one-carbon unit as the sole methyl donor. This study aims to examine the role of dTMP biosynthesis in the development of neural tube in mice by inhibition of TS via a specific inhibitor, raltitrexed (RTX). Pregnant mice were intraperitoneally injected with various doses of RTX on gestational day 7.5, and embryos were examined for the presence of NTDs on gestational day 11.5. TS activity and changes of dUMP and dTMP levels were measured following RTX treatment at the optimal dose. DNA damage was determined by detection of phosphorylated replication protein A2 (RPA2) and γ-H2AX in embryos with NTDs induced by RTX. Besides, apoptosis and proliferation were also analyzed in RTX-treated embryos with NTDs. We found that NTDs were highly occurred by the treatment of RTX at the optimal dose of 11.5 mg/kg b/w. RTX treatment significantly inhibited TS activity. Meanwhile, dTMP was decreased associated with the accumulation of dUMP in RTX-treated embryos. Phosphorylated RPA2 and γ-H2AX were significantly increased in RTX-treated embryos with NTDs compared to control. More apoptosis and decreased proliferation were also found in embryos with NTDs induced by RTX. These results indicate that impairment of dTMP biosynthesis caused by RTX led to the development of NTDs in mice. DNA damage and imbalance between apoptosis and proliferation may be potential mechanisms.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Embrião de Mamíferos/efeitos dos fármacos , Defeitos do Tubo Neural/metabolismo , Quinazolinas/toxicidade , Tiofenos/toxicidade , Animais , Western Blotting , Nucleotídeos de Desoxiuracil/metabolismo , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Feminino , Antagonistas do Ácido Fólico/administração & dosagem , Antagonistas do Ácido Fólico/toxicidade , Idade Gestacional , Histonas/metabolismo , Injeções Intraperitoneais , Masculino , Camundongos Endogâmicos C57BL , Defeitos do Tubo Neural/induzido quimicamente , Defeitos do Tubo Neural/genética , Fosforilação/efeitos dos fármacos , Gravidez , Subunidades Proteicas/metabolismo , Quinazolinas/administração & dosagem , Proteína de Replicação A/metabolismo , Tiofenos/administração & dosagem , Timidina Monofosfato/metabolismo , Timidilato Sintase/antagonistas & inibidores , Timidilato Sintase/metabolismo
9.
J Nutr ; 144(4): 419-24, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24500934

RESUMO

Folate metabolism affects DNA synthesis, methylation, mutation rates, genomic stability, and gene expression, which are altered in colon cancer. Serine hydroxymethyltransferase 1 (SHMT1) regulates thymidylate (dTMP) biosynthesis and uracil accumulation in DNA, and as such affects genome stability. Previously, we showed that decreased SHMT1 expression in Shmt1 knockout mice (Shmt1(-/+)) or its impaired nuclear localization, as occurs in mice over-expressing an Shmt1 transgene (Shmt1(tg+)), results in elevated uracil incorporation into DNA, which could affect colon cancer risk. We used these 2 models to determine the effect of altered SHMT1 expression and localization, and its interaction with folate insufficiency, on azoxymethane (AOM)-induced colon cancer in mice. Shmt1(-/+) and Shmt1(tg+) mice were weaned to a control or folate-and-choline-deficient (FCD) diet and fed the diet for 28 or 32 wk, respectively. At 6 wk of age, mice were injected weekly for 6 wk with 10 mg/kg AOM (w/v in saline). Colon uracil concentrations in nuclear DNA were elevated 2-7 fold in Shmt1(-/+) and Shmt1(tg+) mice. However, colon tumor incidence and numbers were not dependent on SHMT1 expression in Shmt1(-/+) or Shmt1(-/-) mice. The FCD diet reduced tumor load independent of Shmt1 genotype. In contrast, Shmt1(tg+) mice exhibited a 30% reduction in tumor incidence, a 50% reduction in tumor number, and a 60% reduction in tumor load compared with wild-type mice independent of dietary folate intake. Our data indicate that uracil accumulation in DNA does not predict tumor number in AOM-mediated carcinogenesis. Furthermore, enrichment of SHMT1 in the cytoplasm, as observed in Shmt1(tg+) mice, protects against AOM-mediated carcinogenesis independent of its role in nuclear de novo dTMP biosynthesis.


Assuntos
Carcinogênese/metabolismo , Colo/metabolismo , Neoplasias do Colo/metabolismo , DNA/metabolismo , Modelos Animais de Doenças , Ácido Fólico/metabolismo , Timidina Monofosfato/metabolismo , Animais , Azoximetano , Deficiência de Colina/fisiopatologia , Colo/enzimologia , Colo/patologia , Neoplasias do Colo/patologia , Neoplasias do Colo/prevenção & controle , Cruzamentos Genéticos , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Ácido Fólico/efeitos adversos , Deficiência de Ácido Fólico/fisiopatologia , Glicina Hidroximetiltransferase/biossíntese , Glicina Hidroximetiltransferase/genética , Glicina Hidroximetiltransferase/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína-Lisina 6-Oxidase/genética , Proteína-Lisina 6-Oxidase/metabolismo , Distribuição Aleatória , Carga Tumoral , Uracila/metabolismo
10.
Biochemistry ; 52(51): 9167-76, 2013 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-24261692

RESUMO

Catalytic promiscuity, an evolutionary concept, also provides a powerful tool for gaining mechanistic insights into enzymatic reactions. Members of the alkaline phosphatase (AP) superfamily are highly amenable to such investigation, with several members having been shown to exhibit promiscuous activity for the cognate reactions of other superfamily members. Previous work has shown that nucleotide pyrophosphatase/phosphodiesterase (NPP) exhibits a >106-fold preference for the hydrolysis of phosphate diesters over phosphate monoesters, and that the reaction specificity is reduced 10³-fold when the size of the substituent on the transferred phosphoryl group of phosphate diester substrates is reduced to a methyl group. Here we show additional specificity contributions from the binding pocket for this substituent (herein termed the R' substituent) that account for an additional ~250-fold differential specificity with the minimal methyl substituent. Removal of four hydrophobic side chains suggested on the basis of structural inspection to interact favorably with R' substituents decreases phosphate diester reactivity 104-fold with an optimal diester substrate (R' = 5'-deoxythymidine) and 50-fold with a minimal diester substrate (R' = CH3). These mutations also enhance the enzyme's promiscuous phosphate monoesterase activity by nearly an order of magnitude, an effect that is traced by mutation to the reduction of unfavorable interactions with the two residues closest to the nonbridging phosphoryl oxygen atoms. The quadruple R' pocket mutant exhibits the same activity toward phosphate diester and phosphate monoester substrates that have identical leaving groups, with substantial rate enhancements of ~10¹¹-fold. This observation suggests that the Zn²âº bimetallo core of AP superfamily enzymes, which is equipotent in phosphate monoester and diester catalysis, has the potential to become specialized for the hydrolysis of each class of phosphate esters via addition of side chains that interact with the substrate atoms and substituents that project away from the Zn²âº bimetallo core.


Assuntos
Monofosfato de Adenosina/metabolismo , Proteínas de Bactérias/metabolismo , Modelos Moleculares , Diester Fosfórico Hidrolases/metabolismo , Pirofosfatases/metabolismo , Monofosfato de Adenosina/química , Substituição de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Biocatálise , Hidrólise , Interações Hidrofóbicas e Hidrofílicas , Metilação , Conformação Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Nitrofenóis/química , Nitrofenóis/metabolismo , Compostos Organofosforados/química , Compostos Organofosforados/metabolismo , Diester Fosfórico Hidrolases/química , Diester Fosfórico Hidrolases/genética , Mapas de Interação de Proteínas , Pirofosfatases/química , Pirofosfatases/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Timidina Monofosfato/análogos & derivados , Timidina Monofosfato/química , Timidina Monofosfato/metabolismo , Xanthomonas axonopodis/enzimologia , Zinco/química , Zinco/metabolismo
11.
Biol Chem ; 394(2): 261-70, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23087103

RESUMO

Adenosine 3',5'-cyclic monophosphate and guanosine 3',5'-cyclic monophosphate are second messengers that regulate multiple physiological functions. The existence of additional cyclic nucleotides in mammalian cells was postulated many years ago, but technical problems hampered development of the field. Using highly specific and sensitive mass spectrometry methods, soluble guanylyl cyclase has recently been shown to catalyze the formation of several cyclic nucleotides in vitro. This minireview discusses the broad substrate-specificity of soluble guanylyl cyclase and the possible second messenger roles of cyclic nucleotides other than adenosine 3',5'-cyclic monophosphate and guanosine 3',5'-cyclic monophosphate. We hope that this article stimulates productive and critical research in an area that has been neglected for many years.


Assuntos
CMP Cíclico/metabolismo , IMP Cíclico/metabolismo , Guanilato Ciclase/metabolismo , Nucleotídeos Cíclicos/metabolismo , Nucleotídeos de Purina/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Sistemas do Segundo Mensageiro , Timidina Monofosfato/metabolismo , Uridina Monofosfato/metabolismo , Animais , Guanilato Ciclase/química , Humanos , Modelos Biológicos , Receptores Citoplasmáticos e Nucleares/química , Guanilil Ciclase Solúvel
12.
J Biol Chem ; 286(39): 33872-8, 2011 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-21832075

RESUMO

Development of tumor-specific probes for imaging by positron emission tomography has broad implications in clinical oncology, such as diagnosis, staging, and monitoring therapeutic responses in patients, as well as in biomedical research. Thymidylate synthase (TSase)-based de novo biosynthesis of DNA is an important target for drug development. Increased DNA replication in proliferating cancerous cells requires TSase activity, which catalyzes the reductive methylation of dUMP to dTMP using (R)-N(5),N(10)-methylene-5,6,7,8-tetrahydrofolate (MTHF) as a cofactor. In principle, radiolabeled MTHF can be used as a substrate for this reaction to identify rapidly dividing cells. In this proof-of-principle study, actively growing (log phase) breast cancer (MCF7, MDA-MB-231, and hTERT-HME1), normal breast (human mammary epithelial and MCF10A), colon cancer (HT-29), and normal colon (FHC) cells were incubated with [(14)C]MTHF in culture medium from 30 min to 2 h, and uptake of radiotracer was measured. Cancerous cell lines incorporated significantly more radioactivity than their normal counterparts. The uptake of radioactively labeled MTHF depended upon a combination of cell doubling time, folate receptor status, S phase percentage, and TSase expression in the cells. These findings suggest that the recently synthesized [(11)C]MTHF may serve as a new positron emission tomography tracer for cancer imaging.


Assuntos
Replicação do DNA , DNA de Neoplasias/biossíntese , Neoplasias , Tomografia por Emissão de Pósitrons/métodos , Traçadores Radioativos , Tetra-Hidrofolatos/metabolismo , Divisão Celular , Linhagem Celular Tumoral , Nucleotídeos de Desoxiuracil/metabolismo , Humanos , Metilação , Proteínas de Neoplasias/metabolismo , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Radiografia , Timidina Monofosfato/metabolismo , Timidilato Sintase/metabolismo
13.
J Virol ; 85(2): 957-67, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21068232

RESUMO

The origin-specific replication of the herpes simplex virus 1 genome requires seven proteins: the helicase-primase (UL5-UL8-UL52), the DNA polymerase (UL30-UL42), the single-strand DNA binding protein (ICP8), and the origin-binding protein (UL9). We reconstituted these proteins, excluding UL9, on synthetic minicircular DNA templates and monitored leading and lagging strand DNA synthesis using the strand-specific incorporation of dTMP and dAMP. Critical features of the assays that led to efficient leading and lagging stand synthesis included high helicase-primase concentrations and a lagging strand template whose sequence resembled that of the viral DNA. Depending on the nature of the minicircle template, the replication complex synthesized leading and lagging strand products at molar ratios varying between 1:1 and 3:1. Lagging strand products (∼0.2 to 0.6 kb) were significantly shorter than leading strand products (∼2 to 10 kb), and conditions that stimulated primer synthesis led to shorter lagging strand products. ICP8 was not essential; however, its presence stimulated DNA synthesis and increased the length of both leading and lagging strand products. Curiously, human DNA polymerase α (p70-p180 or p49-p58-p70-p180), which improves the utilization of RNA primers synthesized by herpesvirus primase on linear DNA templates, had no effect on the replication of the minicircles. The lack of stimulation by polymerase α suggests the existence of a macromolecular assembly that enhances the utilization of RNA primers and may functionally couple leading and lagging strand synthesis. Evidence for functional coupling is further provided by our observations that (i) leading and lagging strand synthesis produce equal amounts of DNA, (ii) leading strand synthesis proceeds faster under conditions that disable primer synthesis on the lagging strand, and (iii) conditions that accelerate helicase-catalyzed DNA unwinding stimulate decoupled leading strand synthesis but not coordinated leading and lagging strand synthesis.


Assuntos
Replicação do DNA , DNA Circular/metabolismo , Herpesvirus Humano 1/enzimologia , Proteínas Virais/metabolismo , Primers do DNA/genética , DNA Circular/genética , DNA Viral/genética , DNA Viral/metabolismo , Nucleotídeos de Desoxiadenina/metabolismo , Moldes Genéticos , Timidina Monofosfato/metabolismo , Proteínas Virais/isolamento & purificação
15.
Chirality ; 24(12): 977-86, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23001645

RESUMO

Novel chiral Schiff base ligands (R)/(S)-2-amino-3-(((1-hydroxypropan-2-yl)imino)methyl)-4H-chromen-4-one (L(1) and L(2)) derived from 2-amino-3-formylchromone and (R/S)-2-amino-1-propanol and their Cu(II)/Zn(II) complexes (R1, S1, R2, and S2) were synthesized. The complexes were characterized by elemental analysis, infrared (IR), hydrogen ((1) H) and carbon ((13)C) nuclear magnetic resonance (NMR), electrospray ionization-mass spectra (ESI-MS), and molar conductance measurements. The DNA binding studies of the complexes with calf thymus were carried out by employing different biophysical methods and molecular docking studies that revealed that complexes R1 and S1 prefers the guanine-cytosine-rich region, whereas R2 and prefers the adenine-thymine residues in the major groove of DNA. The relative trend in K(b) values followed the order R1>S1>R2>S2. This observation together with the findings of circular dichroic and fluorescence studies revealed maximal potential of (R)-enantiomeric form of complexes to bind DNA. Furthermore, the absorption studies with mononucleotides were also monitored to examine the base-specific interactions of the complexes that revealed a higher propensity of Cu(II) complexes for guanosine-5'-monophosphate disodium salt, whereas Zn(II) complexes preferentially bind to thymidine-5'-monophosphate disodium salt. The cleavage activity of R1 and R2 with pBR322 plasmid DNA was examined by gel electrophoresis that revealed that they are good DNA cleavage agents; nevertheless, R1 proved to show better DNA cleavage ability. Topoisomerase II inhibitory activity of complex R1 revealed that the complex inhibits topoisomerase II catalytic activity at a very low concentration (25 µM). Furthermore, in vitro antitumor activity of complexes R1 and S1 were screened against human carcinoma cell lines of different histological origin.


Assuntos
Cromonas/química , Cobre/química , DNA/metabolismo , Compostos Organometálicos/química , Compostos Organometálicos/metabolismo , Zinco/química , Absorção , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Sítios de Ligação , Linhagem Celular Tumoral , DNA/química , DNA Topoisomerases Tipo II/metabolismo , Desenho de Fármacos , Bócio Subesternal , Guanosina Monofosfato/metabolismo , Humanos , Modelos Moleculares , Conformação de Ácido Nucleico , Compostos Organometálicos/síntese química , Compostos Organometálicos/farmacologia , Estereoisomerismo , Especificidade por Substrato , Timidina Monofosfato/metabolismo , Viscosidade
16.
FEBS J ; 289(6): 1625-1649, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34694685

RESUMO

De novo thymidylate synthesis is a crucial pathway for normal and cancer cells. Deoxythymidine monophosphate (dTMP) is synthesized by the combined action of three enzymes: serine hydroxymethyltransferase (SHMT1), dihydrofolate reductase (DHFR) and thymidylate synthase (TYMS), with the latter two being targets of widely used chemotherapeutics such as antifolates and 5-fluorouracil. These proteins translocate to the nucleus after SUMOylation and are suggested to assemble in this compartment into the thymidylate synthesis complex. We report the intracellular dynamics of the complex in cancer cells by an in situ proximity ligation assay, showing that it is also detected in the cytoplasm. This result indicates that the role of the thymidylate synthesis complex assembly may go beyond dTMP synthesis. We have successfully assembled the dTMP synthesis complex in vitro, employing tetrameric SHMT1 and a bifunctional chimeric enzyme comprising human thymidylate synthase and dihydrofolate reductase. We show that the SHMT1 tetrameric state is required for efficient complex assembly, indicating that this aggregation state is evolutionarily selected in eukaryotes to optimize protein-protein interactions. Lastly, our results regarding the activity of the complete thymidylate cycle in vitro may provide a useful tool with respect to developing drugs targeting the entire complex instead of the individual components.


Assuntos
Timidina Monofosfato , Timidilato Sintase , Núcleo Celular/metabolismo , Glicina Hidroximetiltransferase/genética , Glicina Hidroximetiltransferase/metabolismo , Humanos , Tetra-Hidrofolato Desidrogenase/genética , Tetra-Hidrofolato Desidrogenase/metabolismo , Timidina Monofosfato/metabolismo , Timidilato Sintase/genética , Timidilato Sintase/metabolismo
17.
Anal Biochem ; 416(1): 112-6, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21620793

RESUMO

Tyrosyl DNA phosphodiesterase 2 (TDP2), a newly discovered enzyme that cleaves 5'-phosphotyrosyl bonds, is a potential target for chemotherapy. TDP2 possesses both 3'- and 5'-tyrosyl-DNA phosphodiesterase activity, which is generally measured in a gel-based assay using 3'- and 5'-phosphotyrosyl linkage at the 3' and 5' ends of an oligonucleotide. To understand the enzymatic mechanism of this novel enzyme, the gel-based assay is useful, but this technique is cumbersome for TDP2 inhibitor screening. For this reason, we have designed a novel assay using p-nitrophenyl-thymidine-5'-phosphate (T5PNP) as a substrate. This assay can be used in continuous colorimetric assays in a 96-well format. We compared the salt and pH effect on product formation with the colorimetric and gel-based assays and showed that they behave similarly. Steady-state kinetic studies showed that the 5' activity of TDP2 is 1000-fold more efficient than T5PNP. Tyrosyl DNA phosphodiesterase 1 (TDP1) and human AP-endonuclease 1 (APE1) could not hydrolyze T5PNP. Sodium orthovanadate, a known inhibitor of TDP2, inhibits product formation from T5PNP by TDP2 (IC(50)=40 mM). Our results suggest that this novel assay system with this new TDP2 substrate can be used for inhibitor screening in a high-throughput manner.


Assuntos
Colorimetria/métodos , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Ligação a DNA , Humanos , Proteínas Nucleares/antagonistas & inibidores , Diester Fosfórico Hidrolases , Especificidade por Substrato , Timidina Monofosfato/análogos & derivados , Timidina Monofosfato/química , Timidina Monofosfato/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Vanadatos/farmacologia
18.
Org Biomol Chem ; 9(6): 1799-808, 2011 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-21264378

RESUMO

Ravidomycin V and related compounds, e.g., FE35A-B, exhibit potent anticancer activities against various cancer cell lines in the presence of visible light. The amino sugar moieties (D-ravidosamine and its analogues, respectively) in these molecules contribute to the higher potencies of ravidomycin and analogues when compared to closely related compounds with neutral or branched sugars. Within the ravidomycin V biosynthetic gene cluster, five putative genes encoding NDP-D-ravidosamine biosynthetic enzymes were identified. Through the activities of the isolated enzymes in vitro, it is demonstrated that ravD, ravE, ravIM, ravAMT and ravNMT encode TDP-D-glucose synthase, TDP-4-keto-6-deoxy-D-glucose-4,6-dehydratase, TDP-4-keto-6-deoxy-D-glucose-3,4-ketoisomerase, TDP-3-keto-6-deoxy-D-galactose-3-aminotransferase, and TDP-3-amino-3,6-dideoxy-D-galactose-N,N-dimethyl-transferase, respectively. A protocol for a one-pot enzymatic synthesis of TDP-D-ravidosamine has been developed. The results presented here now set the stage to produce TDP-D-ravidosamine routinely for glycosylation studies.


Assuntos
Bacillales/enzimologia , Escherichia coli/enzimologia , Glucofosfatos/química , Açúcares de Nucleosídeo Difosfato/química , Salmonella typhimurium/enzimologia , Timidina Monofosfato/química , Nucleotídeos de Timina/química , Glucofosfatos/biossíntese , Estrutura Molecular , Família Multigênica , Açúcares de Nucleosídeo Difosfato/metabolismo , Timidina Monofosfato/metabolismo
19.
Bioorg Med Chem ; 19(24): 7603-11, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22061826

RESUMO

We report on Mycobacterium tuberculosis thymidine monophosphate kinase (TMPKmt) inhibitory activities of a series of new 3'- and 5'-modified thymidine analogues including α- and ß-derivatives. In addition, several analogues were synthesized in which the 4-oxygen was replaced by a more lipophilic sulfur atom to probe the influence of this modification on TMPKmt inhibitory activity. Several compounds showed an inhibitory potency in the low micromolar range, with the 5'-arylthiourea 4-thio-α-thymidine analogue being the most active one (K(i)=0.17 µM). This compound was capable of inhibiting mycobacteria growth at a concentration of 25 µg/mL.


Assuntos
Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/enzimologia , Núcleosídeo-Fosfato Quinase/antagonistas & inibidores , Inibidores de Proteínas Quinases/análogos & derivados , Inibidores de Proteínas Quinases/farmacologia , Timidina/análogos & derivados , Timidina/farmacologia , Humanos , Modelos Moleculares , Mycobacterium tuberculosis/crescimento & desenvolvimento , Núcleosídeo-Fosfato Quinase/química , Núcleosídeo-Fosfato Quinase/metabolismo , Inibidores de Proteínas Quinases/síntese química , Timidina/síntese química , Timidina Monofosfato/metabolismo , Tuberculose/tratamento farmacológico
20.
Proteins ; 78(11): 2459-68, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20602460

RESUMO

Thymidine-3'-monophosphate (3'-TMP) is a competitive inhibitor analogue of the 3'-CMP and 3'-UMP natural product inhibitors of bovine pancreatic ribonuclease A (RNase A). Isothermal titration calorimetry experiments show that 3'-TMP binds the enzyme with a dissociation constant (K(d)) of 15 microM making it one of the strongest binding members of the five natural bases found in nucleic acids (A, C, G, T, and U). To further investigate the molecular properties of this potent natural affinity, we have determined the crystal structure of bovine pancreatic RNase A in complex with 3'-TMP at 1.55 A resolution and we have performed NMR binding experiments with 3'-CMP and 3'-TMP. Our results show that binding of 3'-TMP is very similar to other natural and non-natural pyrimidine ligands, demonstrating that single nucleotide affinity is independent of the presence or absence of a 2'-hydroxyl on the ribose moiety of pyrimidines and suggesting that the pyrimidine binding subsite of RNase A is not a significant contributor of inhibitor discrimination. Accumulating evidence suggests that very subtle structural, chemical, and potentially motional variations contribute to ligand discrimination in this enzyme.


Assuntos
Ribonuclease Pancreático/química , Timidina Monofosfato/química , Animais , Bovinos , Cristalografia por Raios X , Monofosfato de Citidina/química , Monofosfato de Citidina/metabolismo , Humanos , Ligantes , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Ribonuclease Pancreático/metabolismo , Termodinâmica , Timidina Monofosfato/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA