Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Immunohorizons ; 3(8): 412-421, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31455692

ABSTRACT

Infection with the intestinal parasite Giardia duodenalis is one of the most common causes of diarrheal disease in the world. Previous work has demonstrated that the cells and mechanisms of the adaptive immune system are critical for clearance of this parasite. However, the innate system has not been as well studied in the context of Giardia infection. We have previously demonstrated that Giardia infection leads to the accumulation of a population of CD11b+, F4/80+, ARG1+, and NOS2+ macrophages in the small intestinal lamina propria. In this report, we sought to identify the accumulation mechanism of duodenal macrophages during Giardia infection and to determine if these cells were essential to the induction of protective Giardia immunity. We show that F4/80+, CD11b+, CD11cint, CX3CR1+, MHC class II+, Ly6C-, ARG1+, and NOS2+ macrophages accumulate in the small intestine during infections in mice. Consistent with this resident macrophage phenotype, macrophage accumulation does not require CCR2, and the macrophages incorporate EdU, indicating in situ proliferation rather than the recruitment of monocytes. Depletion of macrophages using anti-CSF1R did not impact parasite clearance nor development of regulatory T cell or Th17 cellular responses, suggesting that these macrophages are dispensable for protective Giardia immunity.


Subject(s)
Giardia lamblia/immunology , Giardiasis/immunology , Macrophages/immunology , Animals , Cell Proliferation/drug effects , Cytokines/genetics , Deoxyuracil Nucleotides/administration & dosage , Deoxyuracil Nucleotides/pharmacology , Duodenum/immunology , Duodenum/parasitology , Gene Knockout Techniques , Giardiasis/parasitology , Intestine, Small/immunology , Macrophages/classification , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/immunology , Phenotype , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Th17 Cells/immunology
2.
Sci Rep ; 9(1): 11798, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409825

ABSTRACT

Acute liver failure (ALF) is characterized by loss of liver function in response to sustained augmentation of the acute-phase response (APR) in the liver, which can progress even to death. Although the inflammatory interleukin-6 (IL-6)-axis is a crucial factor that drives the hepatic APR by releasing diverse acute-phase proteins (APPs), therapeutic strategies to block the IL-6-STAT3-mediated APR are not well developed. Here, we show that the nuclear receptor retinoic acid-related orphan receptor α (RORα) limits APR-mediated liver injury by inhibiting the hepatic IL-6-STAT3 signaling pathway. Administration of JC1-40, an RORα activator, diminished diethylnitrosamine-induced acute liver injury and repressed transcriptional expression of APPs such as CXCL1 and LCN2 in mice. IL-6-mediated activation of STAT3 was repressed after RORα activation by either adenoviral infusion of RORα or JC1-40 treatment in primary hepatocytes. Activation of RORα decreased transcriptional expression of IL-6 receptor α, an upstream activator of STAT3, both in vitro and in vivo. This may be one mechanism underlying the RORα-mediated inhibition of STAT3. Taken together, our results suggest that RORα is a regulator of the hepatic IL-6-STAT3 signaling pathway and may be a new therapeutic target for treating APR-associated inflammatory ALF.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Interleukin-6/genetics , Liver Failure, Acute/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , STAT3 Transcription Factor/genetics , Acute-Phase Reaction/genetics , Adenoviridae/genetics , Animals , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Chemokine CXCL1/genetics , Deoxyuracil Nucleotides/pharmacology , Diethylnitrosamine/toxicity , Disease Models, Animal , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Humans , Hydroxycholesterols/pharmacology , Liver/injuries , Liver/metabolism , Liver/pathology , Liver Failure, Acute/genetics , Liver Failure, Acute/pathology , Mice , Nuclear Receptor Subfamily 1, Group F, Member 1/antagonists & inhibitors , Primary Cell Culture , Signal Transduction/drug effects
3.
J Med Chem ; 62(4): 1859-1874, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30653317

ABSTRACT

Hepatitis C virus (HCV) nucleoside inhibitors display pan-genotypic activity, a high barrier to the selection of resistant virus, and are some of the most potent direct-acting agents with durable sustained virologic response in humans. Herein, we report, the discovery of ß-d-2'-Br,2'-F-uridine phosphoramidate diastereomers 27 and 28, as nontoxic pan-genotypic anti-HCV agents. Extensive profiling of these two phosphorous diastereomers was performed to select one for in-depth preclinical profiling. The 5'-triphosphate formed from these phosphoramidates selectively inhibited HCV NS5B polymerase with no inhibition of human polymerases and cellular mitochondrial RNA polymerase up to 100 µM. Both are nontoxic by a variety of measures and display good stability in human blood and favorable metabolism in human intestinal microsomes and liver microsomes. Ultimately, a preliminary oral pharmacokinetics study in male beagles showed that 28 is superior to 27 and is an attractive candidate for further studies to establish its potential value as a new clinical anti-HCV agent.


Subject(s)
Antiviral Agents/pharmacology , Deoxyribonucleosides/pharmacology , Deoxyuracil Nucleotides/pharmacology , Hepacivirus/drug effects , Prodrugs/pharmacology , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacokinetics , Cell Line, Tumor , Deoxyribonucleosides/chemical synthesis , Deoxyribonucleosides/pharmacokinetics , Deoxyuracil Nucleotides/chemical synthesis , Deoxyuracil Nucleotides/pharmacokinetics , Dogs , Drug Discovery , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Humans , Male , Microsomes, Liver/metabolism , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Viral Nonstructural Proteins/antagonists & inhibitors
4.
Antibiot Khimioter ; 61(11-12): 9-15, 2016.
Article in English, Russian | MEDLINE | ID: mdl-29558055

ABSTRACT

Different phosphocholine-cardiolipin-2'-deoxyuridine inclusion complexes were developed, that allowed to compose a water-soluble form of nucleoside analogues with previously defined antituberculosis activity. It was found that the resulting liposomes effectively penetrated to the cells. The increase of cytotoxicity was undoubtedly indicative of accumulation of the nucleoside in the cell culture. The result proved the ability of the liposomes for delivery of the low-soluble compounds to the cells for further investigation of their efficacy. It was shown that treatment of the bacterial cells with the llposomes of the modified nucleosides did not affect the bacterial growth.


Subject(s)
Antitubercular Agents , Cardiolipins , Deoxyuracil Nucleotides , Mycobacterium smegmatis/growth & development , Mycobacterium tuberculosis/growth & development , Phosphorylcholine , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Antitubercular Agents/pharmacology , Cardiolipins/chemistry , Cardiolipins/pharmacology , Deoxyuracil Nucleotides/chemical synthesis , Deoxyuracil Nucleotides/chemistry , Deoxyuracil Nucleotides/pharmacology , Liposomes , Phosphorylcholine/chemistry , Phosphorylcholine/pharmacology
5.
J Med Chem ; 54(13): 4847-62, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-21657202

ABSTRACT

A series of 5-substituted 2'-deoxyuridine monophosphate analogues has been synthesized and evaluated as potential inhibitors of mycobacterial ThyX, a novel flavin-dependent thymidylate synthase in Mycobacterium tuberculosis. A systematic SAR study led to the identification of compound 5a, displaying an IC(50) value against mycobacterial ThyX of 0.91 µM. This derivative lacks activity against the classical mycobacterial thymidylate synthase ThyA (IC(50) > 50 µM) and represents the first example of a selective mycobacterial FDTS inhibitor.


Subject(s)
Antitubercular Agents/chemical synthesis , Deoxyuracil Nucleotides/chemical synthesis , Flavins/metabolism , Mycobacterium tuberculosis/enzymology , Thymidylate Synthase/antagonists & inhibitors , Antitubercular Agents/chemistry , Deoxyuracil Nucleotides/chemistry , Deoxyuracil Nucleotides/pharmacology , Structure-Activity Relationship , Thymidylate Synthase/chemistry
6.
Antimicrob Agents Chemother ; 54(8): 3187-96, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20516278

ABSTRACT

The hepatitis C virus (HCV) NS5B RNA polymerase facilitates the RNA synthesis step during the HCV replication cycle. Nucleoside analogs targeting the NS5B provide an attractive approach to treating HCV infections because of their high barrier to resistance and pan-genotype activity. PSI-7851, a pronucleotide of beta-D-2'-deoxy-2'-fluoro-2'-C-methyluridine-5'-monophosphate, is a highly active nucleotide analog inhibitor of HCV for which a phase 1b multiple ascending dose study of genotype 1-infected individuals was recently completed (M. Rodriguez-Torres, E. Lawitz, S. Flach, J. M. Denning, E. Albanis, W. T. Symonds, and M. M. Berry, Abstr. 60th Annu. Meet. Am. Assoc. Study Liver Dis., abstr. LB17, 2009). The studies described here characterize the in vitro antiviral activity and cytotoxicity profile of PSI-7851. The 50% effective concentration for PSI-7851 against the genotype 1b replicon was determined to be 0.075+/-0.050 microM (mean+/-standard deviation). PSI-7851 was similarly effective against replicons derived from genotypes 1a, 1b, and 2a and the genotype 1a and 2a infectious virus systems. The active triphosphate, PSI-7409, inhibited recombinant NS5B polymerases from genotypes 1 to 4 with comparable 50% inhibitory concentrations. PSI-7851 is a specific HCV inhibitor, as it lacks antiviral activity against other closely related and unrelated viruses. PSI-7409 also lacked any significant activity against cellular DNA and RNA polymerases. No cytotoxicity, mitochondrial toxicity, or bone marrow toxicity was associated with PSI-7851 at the highest concentration tested (100 microM). Cross-resistance studies using replicon mutants conferring resistance to modified nucleoside analogs showed that PSI-7851 was less active against the S282T replicon mutant, whereas cells expressing a replicon containing the S96T/N142T mutation remained fully susceptible to PSI-7851. Clearance studies using replicon cells demonstrated that PSI-7851 was able to clear cells of HCV replicon RNA and prevent viral rebound.


Subject(s)
Antiviral Agents/pharmacology , Deoxyuracil Nucleotides/pharmacology , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Prodrugs/pharmacology , Virus Replication/drug effects , Amides/chemistry , Amides/pharmacology , Antiviral Agents/chemistry , Cell Line, Tumor , Deoxyuracil Nucleotides/chemistry , Enzyme Inhibitors/chemistry , Genotype , Hepacivirus/classification , Hepacivirus/enzymology , Humans , Phosphoric Acids/chemistry , Phosphoric Acids/pharmacology , Prodrugs/chemistry , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Replicon/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors
7.
Bioorg Med Chem Lett ; 16(20): 5321-3, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16920358

ABSTRACT

Suligovir is a 35-mer homo-oligonucleotide, containing exclusively 4-thio deoxyuridylate, proved to be a potent inhibitor of HIV entry. In this paper, we described the effect of extent of thiolation and the introduction of nuclease-resistant phosphorothioate linkages on the anti-HIV activity of Suligovir. We found that the decreased thiolated nucleotide content decreases the anti-HIV potency of the compound and the introduction of phosphorothioate linkages does not improve its antiviral activity.


Subject(s)
Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/pharmacology , Deoxyuracil Nucleotides/chemical synthesis , HIV/drug effects , Oligonucleotides/chemical synthesis , Oligonucleotides/pharmacology , Thionucleotides/chemistry , Anti-HIV Agents/chemistry , Deoxyuracil Nucleotides/chemistry , Deoxyuracil Nucleotides/pharmacology , Microbial Sensitivity Tests , Molecular Structure , Oligonucleotides/chemistry , Structure-Activity Relationship , Thionucleotides/chemical synthesis , Thionucleotides/pharmacology
8.
J Am Chem Soc ; 127(21): 7729-38, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15913363

ABSTRACT

Ribonucleotide reductases (RNRs) catalyze the conversion of nucleotides to deoxynucleotides providing the monomeric precursors required for DNA replication and repair. The class I RNRs are composed of two homodimeric subunits: R1 and R2. R1 has the active site where nucleotide reduction occurs, and R2 contains the diiron tyrosyl radical (Y*) cofactor essential for radical initiation on R1. Mechanism-based inhibitors, such as 2'-azido-2'-deoxyuridine-5'-diphosphate (N(3)UDP), have provided much insight into the reduction mechanism. N(3)UDP is a stoichiometric inactivator that, upon interaction with RNR, results in loss of the Y* in R2 and formation of a nitrogen-centered radical (N*) covalently attached to C225 (R-S-N*-X) in the active site of R1. N(2) is lost prior to N* formation, and after its formation, stoichiometric amounts of 2-methylene-3-furanone, pyrophosphate, and uracil are also generated. On the basis of the hyperfine interactions associated with N*, it was proposed that N* is also covalently attached to the nucleotide through either the oxygen of the 3'-OH (R-S-N*-O-R') or the 3'-C (R-S-N*-C-OH). To distinguish between the proposed structures, the inactivation was carried out with 3'-[(17)O]-N(3)UDP and N* was examined by 9 and 140 GHz EPR spectroscopy. Broadening of the N* signal was detected and the spectrum simulated to obtain the [(17)O] hyperfine tensor. DFT calculations were employed to determine which structures are in best agreement with the simulated hyperfine tensor and our previous ESEEM data. The results are most consistent with the R-S-N*-C-OH structure and provide evidence for the trapping of a 3'-ketonucleotide in the reduction process.


Subject(s)
Azides/chemistry , Azides/pharmacology , Deoxyuracil Nucleotides/chemistry , Deoxyuracil Nucleotides/pharmacology , Escherichia coli/enzymology , Nucleotides/chemistry , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/chemistry , Electron Spin Resonance Spectroscopy , Enzyme Activation , Models, Molecular , Nucleotides/metabolism , Quantum Theory , Ribonucleotide Reductases/metabolism
9.
Virology ; 334(2): 214-23, 2005 Apr 10.
Article in English | MEDLINE | ID: mdl-15780871

ABSTRACT

We have previously reported the potent in vitro HIV-1 anti-reverse transcriptase activity of a 35-mer of 4-thio-deoxyuridylate [(s(4)dU)(35)]. In efforts to define its activity in a more physiological system, studies were carried out to determine the stage of viral infection that this compound mediates its anti-viral effect. Results of the studies reported herein show that (s(4)dU)(35) is nontoxic and is capable of inhibiting both single and multi-drug resistant HIV strains (IC(50): 0.8-25.4 microg/ml) in vitro. Besides its previously reported anti-RT activity, (s(4)dU)(35) mediated its antiviral action by preventing virus attachment (IC(50): 0.002-0.003 microg/ml), and was stable in vitro and slowly degraded by DNAses. Competition studies and fluorescence resonance energy transfer (FRET) experiments indicated that (s(4)dU)(35) preferentially binds to CD4 receptors, but not to CD48. Confocal laser scanning microscopy (CLSM) studies showed that (s(4)dU)(35) did not penetrate into the cells and colocalized with cell surface thioredoxin. Our studies identify (s(4)dU)(35) as a potential novel HIV entry inhibitor that may have utility as either a systemic antiretroviral or as a preventing agent for HIV transmission.


Subject(s)
Anti-HIV Agents/pharmacology , Deoxyuracil Nucleotides/pharmacology , HIV-1/drug effects , HIV-1/pathogenicity , Reverse Transcriptase Inhibitors/pharmacology , Thionucleotides/pharmacology , Anti-HIV Agents/toxicity , CD4 Antigens/metabolism , Cell Line , Deoxyuracil Nucleotides/chemical synthesis , Deoxyuracil Nucleotides/toxicity , Fluorescence Resonance Energy Transfer , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , HeLa Cells , Humans , Microbial Sensitivity Tests/methods , Microscopy, Confocal , Reverse Transcriptase Inhibitors/toxicity , Thionucleotides/chemical synthesis , Thionucleotides/toxicity
10.
Nucleic Acids Res ; 32(9): 2957-65, 2004.
Article in English | MEDLINE | ID: mdl-15161959

ABSTRACT

The early development of amphibians takes place in the absence of significant transcription and is controlled at the post-transcriptional level. We have reported that in vitro synthesized transcripts injected into axolotl fertilized eggs or oocytes were not continuously degraded as their abundance apparently fluctuated over time, with detected amounts sometimes higher than initial injected amounts. To further characterize this phenomenon, we have co-injected RNA chain terminators to prevent RNA synthesis. This led to the suppression of fluctuations and to a regular decrease in the amount of transcripts that appeared to be more stable in the presence of inhibitors. These observations indicate a coupling between RNA synthesis and an accelerated degradation. Throughout the time course, cRNA molecules could be detected, and their abundance increased in the early phase of the kinetics, supporting the implication of an RNA-dependent RNA polymerase in an asymmetric amplification process. Finally, when the fate of the injected transcripts was investigated in individual oocytes, we observed an absolute increase in abundance in some but not all oocytes, supporting the existence of a limiting step in the initiation of the RNA amplification stochastic process.


Subject(s)
Ambystoma mexicanum/metabolism , Oocytes/metabolism , RNA Stability , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Transcription, Genetic , Zebrafish Proteins , Animals , Deoxyadenosines/pharmacology , Deoxyuracil Nucleotides/pharmacology , Female , Genes, myc/genetics , Kinetics , Oocytes/drug effects , Proto-Oncogene Proteins/genetics , RNA Stability/drug effects , RNA, Complementary/biosynthesis , RNA, Complementary/genetics , RNA, Complementary/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA-Dependent RNA Polymerase/metabolism , Stochastic Processes , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Wnt Proteins , Xenopus/genetics
11.
Cytogenet Genome Res ; 104(1-4): 304-9, 2004.
Article in English | MEDLINE | ID: mdl-15162056

ABSTRACT

The DNA lesions responsible for the formation of sister chromatid exchanges (SCEs) have been the object of research for a long time. SCEs can be visualized by growing cells for either two rounds of replication in the presence of 5-bromo-2'-deoxyuridine (BrdU) or for one round with BrdU and the next without. If BrdU is added after cells were treated with a DNA-damaging agent, the effect on SCEs can only be analyzed in the second post-treatment mitosis. If one wishes to analyze the first post-treatment mitosis, cells unifilarily labeled with BrdU must be treated. Due to the highly reactive bromine atom, BrdU interacts with such agents like ionizing and UV radiation enhancing the frequency of SCEs. However, its precise role in this process was difficult to assess for a long time, because no alternative technique existed that allowed differential staining of chromatids. We have recently developed a method to differentially label sister chromatids with biotin-16-2'-deoxyuridine-5'-triphosphate (biotin-dUTP) circumventing the disadvantage of BrdU. This technique was applied to study the SCEs induced by ionizing and UV radiation as well as by mitomycin C, DNaseI and AluI. This article is a review of the results and conclusions of our previous studies.


Subject(s)
Biotin/analogs & derivatives , Chromosomes/radiation effects , Sister Chromatid Exchange , Animals , Biotin/pharmacology , Bromodeoxyuridine/pharmacology , Bromodeoxyuridine/toxicity , CHO Cells/drug effects , CHO Cells/radiation effects , CHO Cells/ultrastructure , Chromosome Inversion , Chromosomes/drug effects , Cricetinae , Cricetulus , Cross-Linking Reagents/pharmacology , Cross-Linking Reagents/toxicity , DNA Damage , DNA Replication , Deoxyuracil Nucleotides/pharmacology , Free Radicals , G1 Phase , Humans , Mitomycin/pharmacology , Mitomycin/toxicity , Radiation-Sensitizing Agents/pharmacology , Sister Chromatid Exchange/drug effects , Sister Chromatid Exchange/physiology , Sister Chromatid Exchange/radiation effects , Staining and Labeling , Ultraviolet Rays/adverse effects , X-Rays/adverse effects
12.
Biochem Pharmacol ; 66(3): 471-9, 2003 Aug 01.
Article in English | MEDLINE | ID: mdl-12907246

ABSTRACT

Nucleoside analogs act as prodrugs that must be converted to 5'-phosphates by intracellular kinases to become active in the treatment of viral and oncological diseases. Activation may be reversed by dephosphorylation if the 5'-phosphates are substrates for 5'-nucleotidases. Dephosphorylation by cytosolic enzymes decreases the efficacy of the analogs, whereas dephosphorylation by mitochondrial enzymes may decrease mitochondrial toxicity. Both effects may influence the outcome of therapy. We investigated the dephosphorylation of the 5'-phosphates of commonly used nucleoside analogs by two cytosolic (cN-II and dNT-1) and one mitochondrial (dNT-2) nucleotidase. Most uracil/thymine nucleotide analogs were dephosphorylated by all three human enzymes but cytosine-containing nucleotide analogs were inactive. Only cN-II showed some activity with the monophosphates of the two purine analogs 2-chloro-2'-deoxyadenosine and 9-beta-D-arabinosylguanine. We conclude that overproduction of any of the three 5'-nucleotidases cannot explain development of resistance against cytosine analogs but that overproduction of cN-II could lead to resistance against purine analogs. Of the tested analogs, only (E)-5-(2-bromovinyl)-2'-deoxyuridine was preferentially dephosphorylated by mitochondrial dNT-2. We propose that in future developments of analogs this aspect be considered in order to reduce mitochondrial toxicity. We tested inhibition of dNT-1 and dNT-2 by a large variety of synthetic metabolically stable nucleoside phosphonate analogs and found one (PMcP-U) that inhibited dNT-1 and dNT-2 competitively and a second (DPB-T) that inhibited dNT-2 by mixed inhibition. Both inhibitors are useful for specific 5'-nucleotidase assays and structural studies and may open up possibilities for therapy.


Subject(s)
5'-Nucleotidase/metabolism , Mitochondria/enzymology , 5'-Nucleotidase/antagonists & inhibitors , Cytosol/enzymology , Cytosol/metabolism , Deoxyuracil Nucleotides/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Substrate Specificity
13.
J Med Chem ; 45(23): 5157-72, 2002 Nov 07.
Article in English | MEDLINE | ID: mdl-12408726

ABSTRACT

Novel cycloSal-BVDUMP triesters 2-4 5-[(E)-2-bromovinyl]-2'-deoxyuridine (BVDU, 1) have been studied with regard to their potential anti-EBV activity. In addition to the 3'-unmodified cycloSal-BVDUMP triesters 2a-f, the 3'-hydroxyl function has been esterified with different aliphatic carboxylic acids (3a-g) and alpha-amino acids having natural and nonnatural Calpha-configuration (4a-m). In addition to the synthesis of these compounds, different physicochemical properties of the new derivatives will be reported, i.e., lipophilicity and hydrolysis behavior. It could be proven that the monophosphate BVDUMP and not 3',5'-cyclic BVDUMP was delivered from most of the compounds by chemical hydrolysis in phosphate buffers at pH 6.8 and 7.3 as well as P3HR-1 cell extracts. Finally, the new compounds were tested for their anti-EBV activity. As a result, the prototype compounds and particularly triesters 2c,d exhibited pronounced anti-EBV activity making these compounds promising candidates for further development. However, the 3'-ester derivatives were devoid of any antiviral activity while the 3'-aminoacyl derivatives showed an antiviral activity dependent upon the amino acid and the Calpha-configuration


Subject(s)
Antiviral Agents/chemical synthesis , Bromodeoxyuridine/analogs & derivatives , Bromodeoxyuridine/chemical synthesis , Deoxyuracil Nucleotides/chemical synthesis , Herpesvirus 4, Human/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Bromodeoxyuridine/chemistry , Bromodeoxyuridine/pharmacology , Cell Division/drug effects , Cell Extracts , Deoxyuracil Nucleotides/chemistry , Deoxyuracil Nucleotides/pharmacology , Esters , Humans , Hydrolysis , Kinetics , Structure-Activity Relationship , Tumor Cells, Cultured
14.
J Virol ; 76(20): 10530-2, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12239332

ABSTRACT

We demonstrate that human herpesvirus 8, obtained from the lymphoma cell line BC-3 as well as from Kaposi's sarcoma lesions, carries a gene that encodes a functional thymidylate synthase (TS). The particular characteristics of this enzyme are studied and compared to the characteristics of TSs encoded by other organisms.


Subject(s)
Herpesvirus 8, Human/enzymology , Sarcoma, Kaposi/virology , Thymidylate Synthase/metabolism , Amino Acid Sequence , Deoxyuracil Nucleotides/pharmacology , Enzyme Inhibitors/pharmacology , Fluorodeoxyuridylate/pharmacology , Genes, Viral , Herpesvirus 3, Human/enzymology , Herpesvirus 3, Human/genetics , Herpesvirus 8, Human/genetics , Humans , Lymphoma , Molecular Sequence Data , Sarcoma, Kaposi/pathology , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/genetics , Tumor Cells, Cultured
15.
J Cell Sci ; 115(Pt 15): 3159-69, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12118071

ABSTRACT

The response to DNA damage was analyzed using a cell-free system consisting of Xenopus egg extract and demembranated sperm nuclei. In the absence of DNA-damaging agents, detergent-resistant accumulation of replication protein A appeared in nuclei after a 30 minute incubation, and a considerable portion of the replication protein A signals disappeared during a further 30 minute incubation. Similar replication protein A accumulation was observed in the nuclei after a 30 minute incubation in the extract containing camptothecin, whereas a further 30 minute incubation generated discrete replication protein A foci. The addition of camptothecin also induced formation of gamma-H2AX foci, which have been previously shown to localize at sites of DSBs. Analysis of the time course of DNA replication and results obtained using geminin, an inhibitor of licensing for DNA replication, suggest that the discrete replication protein A foci formed in response to camptothecin-induced DNA damage occur in a DNA-replication-dependent manner. When the nuclei were incubated in the extract containing EcoRI, discrete replication protein A foci were observed at 30 minutes as well as at 60 and 90 minutes after incubation, and the focus-formation of replication protein A was not sensitive to geminin. DNA replication was almost completely inhibited in the presence of EcoRI and the inhibition was sensitive to caffeine, an inhibitor of ataxia telangiectasia mutated protein (ATM) and ATM- and Rad3-related protein (ATR). However, the focus-formation of replication protein A in the presence of EcoRI was not influenced by caffeine treatment. EcoRI-induced incorporation of biotin-dUTP into chromatin was observed following geminin-mediated inhibition of DNA replication, suggesting that the incorporation was the result of DNA repair. The biotin-dUTP signal co-localized with replication protein A foci and was not significantly suppressed or stimulated by the addition of caffeine.


Subject(s)
Biotin/analogs & derivatives , Cell Nucleus/genetics , Cell-Free System/metabolism , DNA Damage/genetics , DNA Repair/genetics , DNA-Binding Proteins/biosynthesis , Eukaryotic Cells/metabolism , Genes, cdc/physiology , Xenopus Proteins , Animals , Ataxia Telangiectasia Mutated Proteins , Biotin/pharmacology , Caffeine/pharmacology , Camptothecin/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/pharmacology , Cell Extracts , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell-Free System/drug effects , DNA Damage/drug effects , DNA Repair/drug effects , DNA-Binding Proteins/drug effects , Deoxyuracil Nucleotides/pharmacology , Eukaryotic Cells/drug effects , Female , Geminin , Genes, cdc/drug effects , Male , Oocytes , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Replication Protein A , Site-Specific DNA-Methyltransferase (Adenine-Specific)/pharmacology , Spermatozoa , Tumor Suppressor Proteins , Xenopus laevis
16.
Article in English | MEDLINE | ID: mdl-11563134

ABSTRACT

A novel series of 5-propynyl-dUMP derivatives, with a variety of leaving groups on the side-chain, was designed as potential mechanism-based inhibitors of thymidylate synthase (TS), and synthesized from 5-iodo-2'-deoxyuridine by Pd(0)-catalyzed coupling, followed by direct phosphorylation with POCl3. All members of the series inhibited TS competitively with Ki-values of 0.015-18 microM. Analogs with fluorine or imidazole-based leaving groups caused rapid, irreversible inactivation of TS.


Subject(s)
Deoxyuracil Nucleotides/pharmacology , Enzyme Inhibitors/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Alkynes/chemical synthesis , Alkynes/chemistry , Alkynes/pharmacology , Deoxyuracil Nucleotides/chemical synthesis , Deoxyuracil Nucleotides/chemistry , Drug Design , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Kinetics
17.
Chem Biol ; 8(5): 445-57, 2001 May.
Article in English | MEDLINE | ID: mdl-11358692

ABSTRACT

BACKGROUND: Using fixed receptor sites derived from high-resolution crystal structures in structure-based drug design does not properly account for ligand-induced enzyme conformational change and imparts a bias into the discovery and design of novel ligands. We sought to facilitate the design of improved drug leads by defining residues most likely to change conformation, and then defining a minimal manifold of possible conformations of a target site for drug design based on a small number of identified flexible residues. RESULTS: The crystal structure of thymidylate synthase from an important pathogenic target Pneumocystis carinii (PcTS) bound to its substrate and the inhibitor, BW1843U89, is reported here and reveals a new conformation with respect to the structure of PcTS bound to substrate and the more conventional antifolate inhibitor, CB3717. We developed an algorithm for determining which residues provide 'soft spots' in the protein, regions where conformational adaptation suggests possible modifications for a drug lead that may yield higher affinity. Remodeling the active site of thymidylate synthase with new conformations for only three residues that were identified with this algorithm yields scores for ligands that are compatible with experimental kinetic data. CONCLUSIONS: Based on the examination of many protein/ligand complexes, we develop an algorithm (SOFTSPOTS) for identifying regions of a protein target that are more likely to accommodate plastically to regions of a drug molecule. Using these indicators we develop a second algorithm (PLASTIC) that provides a minimal manifold of possible conformations of a protein target for drug design, reducing the bias in structure-based drug design imparted by structures of enzymes co-crystallized with inhibitors.


Subject(s)
Algorithms , Drug Design , Membrane Proteins/chemistry , Thymidylate Synthase/chemistry , Amino Acid Motifs/physiology , Binding Sites/physiology , Crystallography/methods , Deoxyuracil Nucleotides/chemistry , Deoxyuracil Nucleotides/metabolism , Deoxyuracil Nucleotides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/metabolism , Folic Acid Antagonists/pharmacology , Ligands , Pliability , Pneumocystis/enzymology , Protein Binding/physiology , Protein Conformation/drug effects , Substrate Specificity , Thymidylate Synthase/metabolism
19.
Biochemistry (Mosc) ; 65(2): 244-9, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10713555

ABSTRACT

The thermostable DNA-polymerase from Thermus thermophilus B35 (Tte-polymerase) was affinity labeled by a binary system of photoreagents comprising base-substituted TTP analogs. The 5;-[32P]-labeled primer was elongated by Tte-polymerase in the presence of a TTP analog containing the photoreactive 2,3,5, 6-tetrafluoro-4-azidobenzoyl group (FAB-4-dUTP). Then the reaction mixture was UV-irradiated (365-450 nm) in the presence or the absence of a photosensitizer (TTP analog containing a pyrene moiety, Pyr-dUTP). The initial rate of the Pyr-dUTP-sensitized photomodification was almost 10-fold higher than the rate of direct photomodification (in the absence of Pyr-dUTP); in the case of the sensitized modification, the product of covalent cross-linking of the photoreactive primer with Tte-polymerase was apparently homogenous according to the data of electrophoresis. The enzyme was protected from the photosensitized modification by dNTP. To confirm the selectivity of the photosensitized modification of Tte-polymerase, another DNA-binding protein (human replication factor A, RPA) was added to the reaction mixture. In the presence of the photosensitizer (Pyr-dUTP), RPA was not labeled and only Tte-polymerase was modified, whereas in the case of direct modification, Tte-polymerase and the p32 and p70 subunits of RPA were labeled. The suggested method enables highly selective affinity modification of DNA-polymerases.


Subject(s)
Affinity Labels , DNA-Directed DNA Polymerase/metabolism , Thermus thermophilus/enzymology , Base Sequence , DNA Primers/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Directed DNA Polymerase/chemistry , Deoxyuracil Nucleotides/pharmacology , Humans , In Vitro Techniques , Photosensitizing Agents/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Replication Protein A , Thymine Nucleotides/pharmacology
20.
Mol Pharmacol ; 57(2): 359-66, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10648646

ABSTRACT

Drug-resistant variants of thymidylate synthase (TS) can potentially be used in gene therapy applications to decrease the myelosuppressive side effects of TS-directed anticancer agents or to select genetically modified cells in vivo. Mutations of proline 303 of human TS confer resistance to TS-directed fluoropyrimidines and antifolates (). We generated the corresponding variants in Escherichia coli TS (ecTS), position 254, to better understand the mechanism by which mutations at this residue confer resistance. In addition, because ecTS is intrinsically resistant to several antifolates when compared with human TS, we suspected that greater resistance could be achieved with the bacterial enzyme. The P254L enzyme conferred >100-fold resistance to both raltitrexed and 5-fluoro-2'-deoxyuridine (FdUrd) compared with wild-type ecTS. Four additional mutants (P254F, P254S, P254G, and P254D), each of which complemented growth of a TS-deficient cell line, were generated, isolated, and characterized. Steady-state values of K(m) for dUMP and k(cat) were not substantially different among the variants and were comparable with the wild-type values, but K(m) for methylenetetrahydrofolate (CH(2)H(4)PteGlu) was >10-fold higher for P254D. Values of k(on) and k(off) for nucleotide binding, which were obtained by stopped-flow spectroscopy, were virtually unchanged among the mutants. Drastic differences were observed for CH(2)H(4)PteGlu binding, with K(d) values >15-fold higher than observed with the wild-type enzyme; surprisingly, the proposed isomerization reaction that is very evident for the wild-type enzyme is not observed with P254S. The decrease in affinity for CH(2)H(4)PteGlu correlates well with K(i) values obtained for three TS-directed inhibitors. These results show that mutations at Pro-254 specifically affect the initial binding interactions between enzyme and cofactor and also alter the ability of the mutant enzymes to undergo conformational changes that occur on ternary complex formation. The crystal structure of P254S was determined at 1.5 A resolution and is the most precise structure of TS available. When compared with wild-type TS, the structure shows local conformational changes affecting mostly Asp-253; its carbonyl is rotated approximately 40 degrees, and the side chain forms an ion pair with Arg-225.


Subject(s)
Escherichia coli/enzymology , Folic Acid Antagonists/pharmacology , Thymidylate Synthase/metabolism , Amino Acid Substitution , Crystallography, X-Ray , Deoxyuracil Nucleotides/pharmacology , Drug Resistance , Drug Resistance, Microbial/physiology , Enzyme Inhibitors/pharmacology , Escherichia coli/drug effects , Escherichia coli/genetics , Fluorodeoxyuridylate/pharmacology , Humans , Kinetics , Mutation , Proline/metabolism , Protein Conformation , Quinazolines/pharmacology , Tetrahydrofolates/pharmacology , Thymidylate Synthase/antagonists & inhibitors , Thymidylate Synthase/chemistry , Thymidylate Synthase/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL