Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Commun Biol ; 5(1): 547, 2022 06 06.
Article in English | MEDLINE | ID: mdl-35668202

ABSTRACT

Blocking Plasmodium falciparum human-to-mosquito transmission is essential for malaria elimination, nonetheless drugs killing the pathogenic asexual stages are generally inactive on the parasite transmissible stages, the gametocytes. Due to technical and biological limitations in high throughput screening of non-proliferative stages, the search for gametocyte-killing molecules so far tested one tenth the number of compounds screened on asexual stages. Here we overcome these limitations and rapidly screened around 120,000 compounds, using not purified, bioluminescent mature gametocytes. Orthogonal gametocyte assays, selectivity assays on human cells and asexual parasites, followed by compound clustering, brought to the identification of 84 hits, half of which are gametocyte selective and half with comparable activity against sexual and asexual parasites. We validated seven chemotypes, three of which are, to the best of our knowledge, novel. These molecules are able to inhibit male gametocyte exflagellation and block parasite transmission through the Anopheles mosquito vector in a standard membrane feeding assay. This work shows that interrogating a wide and diverse chemical space, with a streamlined gametocyte HTS and hit validation funnel, holds promise for the identification of dual stage and gametocyte-selective compounds to be developed into new generation of transmission blocking drugs for malaria elimination.


Subject(s)
Anopheles , Malaria , Animals , High-Throughput Screening Assays , Humans , Male , Plasmodium falciparum
2.
Bioorg Med Chem ; 57: 116631, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35123179

ABSTRACT

Zika virus (ZIKV) is a member of the Flaviviridae family that can cause neurological disorders and congenital malformations. The NS2B-NS3 viral serine protease is an attractive target for the development of new antiviral agents against ZIKV. We report here a SAR study on a series of substrate-like linear tripeptides that inhibit in a non-covalent manner the NS2B-NS3 protease. Optimization of the residues at positions P1, P2, P3 and of the N-terminal and C-terminal portions of the tripeptide allowed the identification of inhibitors with sub-micromolar potency with phenylglycine as arginine-mimicking group and benzylamide as C-terminal fragment. Further SAR exploration and application of these structural changes to a series of peptides having a 4-substituted phenylglycine residue at the P1 position led to potent compounds showing double digit nanomolar inhibition of the Zika protease (IC50 = 30 nM) with high selectivity against trypsin-like proteases and the proteases of other flavivirus, such as Dengue 2 virus (DEN2V) and West Nile virus (WNV).


Subject(s)
Antiviral Agents/pharmacology , Peptides/pharmacology , Protease Inhibitors/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Zika Virus/drug effects , Antiviral Agents/chemical synthesis , Antiviral Agents/chemistry , Dengue Virus/drug effects , Dose-Response Relationship, Drug , Humans , Microbial Sensitivity Tests , Molecular Structure , Peptides/chemical synthesis , Peptides/chemistry , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , RNA Helicases/antagonists & inhibitors , RNA Helicases/metabolism , Serine Endopeptidases/metabolism , Structure-Activity Relationship , Viral Nonstructural Proteins/metabolism , West Nile virus/drug effects , Zika Virus/enzymology
3.
PLoS Negl Trop Dis ; 14(5): e0008339, 2020 05.
Article in English | MEDLINE | ID: mdl-32437349

ABSTRACT

Trypanothione reductase (TR) is a key enzyme that catalyzes the reduction of trypanothione, an antioxidant dithiol that protects Trypanosomatid parasites from oxidative stress induced by mammalian host defense systems. TR is considered an attractive target for the development of novel anti-parasitic agents as it is essential for parasite survival but has no close homologue in humans. We report here the identification of spiro-containing derivatives as inhibitors of TR from Trypanosoma brucei (TbTR), the parasite responsible for Human African Trypanosomiasis. The hit series, identified by high throughput screening, was shown to bind TbTR reversibly and to compete with the trypanothione (TS2) substrate. The prototype compound 1 from this series was also found to impede the growth of Trypanosoma brucei parasites in vitro. The X-ray crystal structure of TbTR in complex with compound 1 solved at 1.98 Å allowed the identification of the hydrophobic pocket where the inhibitor binds, placed close to the catalytic histidine (His 461') and lined by Trp21, Val53, Ile106, Tyr110 and Met113. This new inhibitor is specific for TbTR and no activity was detected against the structurally similar human glutathione reductase (hGR). The central spiro scaffold is known to be suitable for brain active compounds in humans thus representing an attractive starting point for the future treatment of the central nervous system stage of T. brucei infections.


Subject(s)
Antiprotozoal Agents/pharmacology , Enzyme Inhibitors/pharmacology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Toluene/analogs & derivatives , Trypanosoma brucei brucei/drug effects , Antiprotozoal Agents/isolation & purification , Binding Sites , Crystallography, X-Ray , Drug Evaluation, Preclinical , Enzyme Inhibitors/isolation & purification , High-Throughput Screening Assays , NADH, NADPH Oxidoreductases/chemistry , Protein Binding , Protein Conformation , Toluene/isolation & purification , Toluene/pharmacology , Trypanosoma brucei brucei/enzymology
4.
Bioorg Med Chem Lett ; 30(12): 127207, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32354566

ABSTRACT

A previous publication from our laboratory reported the identification of a new class of 2-(1H-imidazo-2-yl)piperazines as potent T. brucei growth inhibitors as potential treatment for Human African Trypanosomiasis (HAT). This work describes the structure-activity relationship (SAR) around the hit compound 1, which led to the identification of the optimized compound 18, a single digit nanomolar inhibitor (EC50 7 nM), not cytotoxic and with optimal in vivo profile that made it a suitable candidate for efficacy studies in a mouse model mimicking the second stage of disease.


Subject(s)
Growth Inhibitors/chemistry , Piperazines/chemistry , Trypanocidal Agents/chemistry , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy , Cell Survival/drug effects , Drug Evaluation, Preclinical , Growth Inhibitors/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Isomerism , Morpholines/chemistry , Piperazines/pharmacology , Quinolines/chemistry , Structure-Activity Relationship , Trypanocidal Agents/pharmacology
5.
Bioorg Med Chem Lett ; 30(8): 127052, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32113841

ABSTRACT

The identification of a new series of growth inhibitors of Trypanosoma cruzi, the causative agent of Chagas' disease, is described. In vitro screening of a subset of compounds from our in-house compound collection against the parasite led to the identification of hit compound 1 with low micromolar inhibition of T. cruzi growth. SAR exploration on the hit compound led to the identification of compounds that show nanomolar parasite growth inhibition (T. cruzi EC50 ≤ 100 nM) and no cytotoxicity in human cells (HeLa CC50 > 50 µM). Further investigation identified CYP51 inhibition (compound 11 CYP51 IC50 52 nM) as a possible mechanism of action of this new class of anti-parasitic agents.


Subject(s)
Drug Discovery , Growth Inhibitors/pharmacology , Pyridines/pharmacology , Trypanocidal Agents/pharmacology , Trypanosoma cruzi/drug effects , Dose-Response Relationship, Drug , Growth Inhibitors/chemical synthesis , Growth Inhibitors/chemistry , Humans , Molecular Structure , Parasitic Sensitivity Tests , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry
6.
Parasit Vectors ; 11(1): 668, 2018 Dec 27.
Article in English | MEDLINE | ID: mdl-30587243

ABSTRACT

BACKGROUND: Novel anti-schistosomal multi-stage drugs are needed because only a single drug, praziquantel, is available for the treatment of schistosomiasis and is poorly effective on larval and juvenile stages of the parasite. Schistosomes have a complex life-cycle and multiple developmental stages in the intermediate and definitive hosts. Acetylation and deacetylation of histones play pivotal roles in chromatin structure and in the regulation of transcription in eukaryotic cells. Histone deacetylase (HDAC) inhibitors modulate acetylation of several other proteins localized both in the nucleus and in the cytoplasm and therefore impact on many signaling networks and biological processes. Histone post-translational modifications may provide parasites with the ability to readily adapt to changes in gene expression required for their development and adaptation to the host environment. The aim of the present study was to screen a HDAC class I inhibitor library in order to identify and characterize novel multi-stage hit compounds. METHODS: We used a high-throughput assay based on the quantitation of ATP in the Schistosoma mansoni larval stage (schistosomula) and screened a library of 1500 class I HDAC inhibitors. Subsequently, a few hits were selected and further characterized by viability assays and phenotypic analyses on adult parasites by carmine red and confocal microscopy. RESULTS: Three compounds (SmI-124, SmI-148 and SmI-558) that had an effect on the viability of both the schistosomula larval stage and the adult worm were identified. Treatment with sub-lethal doses of SmI-148 and SmI-558 also decreased egg production. Moreover, treatment of adult parasites with SmI-148, and to a lesser extent Sm-124, was associated with histone hyperacetylation. Finally, SmI-148 and SmI-558 treatments of worm pairs caused a phenotype characterized by defects in the parasite reproductive system, with peculiar features in the ovary. In addition, SmI-558 induced oocyte- and vitelline cell-engulfment and signs of degeneration in the uterus and/or oviduct. CONCLUSIONS: We report the screening of a small HDAC inhibitor library and the identification of three novel compounds which impair viability of the S. mansoni larval stage and adult pairs. These compounds are useful tools for studying deacetylase activity during parasite development and for interfering with egg production. Characterization of their specificity for selected S. mansoni versus human HDAC could provide insights that can be used in optimization and compound design.


Subject(s)
Anthelmintics/administration & dosage , Histone Deacetylase Inhibitors/administration & dosage , Ovum/drug effects , Schistosoma mansoni/drug effects , Schistosoma mansoni/growth & development , Schistosomiasis/drug therapy , Acetylation , Animals , Anthelmintics/chemistry , Female , Helminth Proteins/antagonists & inhibitors , Helminth Proteins/genetics , Helminth Proteins/metabolism , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Life Cycle Stages/drug effects , Male , Mice , Mice, Inbred ICR , Ovum/growth & development , Ovum/metabolism , Schistosoma mansoni/enzymology , Schistosoma mansoni/genetics , Schistosomiasis/parasitology
7.
Bioorg Med Chem Lett ; 28(23-24): 3689-3692, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30482621

ABSTRACT

The identification of a new series of growth inhibitors of Trypanosoma brucei rhodesiense, causative agent of Human African Trypanosomiasis (HAT), is described. A selection of compounds from our in-house compound collection was screened in vitro against the parasite leading to the identification of compounds with nanomolar inhibition of T. brucei growth. Preliminary SAR on the hit compound led to the identification of compound 34 that shows low nanomolar parasite growth inhibition (T. brucei EC50 5 nM), is not cytotoxic (HeLa CC50 > 25,000 nM) and is selective over other parasites, such as Trypanosoma cruzi and Plasmodium falciparum (T. cruzi EC50 8120 nM, P. falciparum EC50 3624 nM).


Subject(s)
Piperazines/chemistry , Piperazines/pharmacology , Trypanocidal Agents/chemistry , Trypanocidal Agents/pharmacology , Trypanosoma brucei brucei/drug effects , Trypanosomiasis, African/drug therapy , HeLa Cells , Humans , Imidazoles/chemistry , Imidazoles/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Structure-Activity Relationship , Trypanosoma brucei brucei/growth & development , Trypanosomiasis, African/parasitology
8.
Bioorg Med Chem Lett ; 28(9): 1540-1544, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29615344

ABSTRACT

Falcipain-2 (FP2) is an essential enzyme in the lifecycle of malaria parasites such as Plasmodium falciparum, and its inhibition is viewed as an attractive mechanism of action for new anti-malarial agents. Selective inhibition of FP2 with respect to a family of human cysteine proteases (that include cathepsins B, K, L and S) is likely to be required for the development of agents targeting FP2. Here we describe a series of P2-modified aminonitrile based inhibitors of FP2 that provide a clear strategy toward addressing selectivity for the P. falciparum and show that it can provide potent FP2 inhibitors with strong selectivity against all four of these human cathepsin isoforms.


Subject(s)
Antimalarials/pharmacology , Cathepsins/antagonists & inhibitors , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Nitriles/pharmacology , Peptidomimetics/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cathepsins/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Malaria, Falciparum/drug therapy , Malaria, Falciparum/metabolism , Molecular Structure , Nitriles/chemical synthesis , Nitriles/chemistry , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Structure-Activity Relationship
9.
ACS Med Chem Lett ; 7(5): 454-9, 2016 May 12.
Article in English | MEDLINE | ID: mdl-27190592

ABSTRACT

The identification of a new series of P. falciparum growth inhibitors is described. Starting from a series of known human class I HDAC inhibitors a SAR exploration based on growth inhibitory activity in parasite and human cells-based assays led to the identification of compounds with submicromolar inhibition of P. falciparum growth (EC50 < 500 nM) and good selectivity over the activity of human HDAC in cells (up to >50-fold). Inhibition of parasital HDACs as the mechanism of action of this new class of selective growth inhibitors is supported by hyperacetylation studies.

10.
Bioorg Med Chem ; 18(16): 6143-8, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20638289

ABSTRACT

A series of ethyl 1H-indole-3-carboxylates 9a(1)(-)(6) and 9b(1)(-)(2) were prepared and evaluated in Huh-7.5 cells. Most of the compounds exhibited anti-hepatitis C virus (HCV) activities at low concentration. The selectivity indices of inhibition on entry and replication of compounds 9a(2) (>10; >16.7) and 9b(1) (>6.25; >16.7) were higher than those of the other evaluated compounds, including the lead compound Arbidol (ARB, 6; 15). Moreover, the selective index of inhibition on entry of compound 9a(3) (>6.25) was higher than that of ARB (6). Of these three initial hits, compound 9a(2) was the most potent.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Indoles/chemistry , Indoles/pharmacology , Antiviral Agents/chemical synthesis , Carboxylic Acids/chemical synthesis , Carboxylic Acids/chemistry , Carboxylic Acids/pharmacology , Cell Line , Hepatitis C/drug therapy , Humans , Indoles/chemical synthesis , Structure-Activity Relationship
11.
J Virol ; 84(1): 34-43, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19828610

ABSTRACT

Scavenger receptor class B type I (SR-BI) is an essential receptor for hepatitis C virus (HCV) and a cell surface high-density-lipoprotein (HDL) receptor. The mechanism of SR-BI-mediated HCV entry, however, is not clearly understood, and the specific protein determinants required for the recognition of the virus envelope are not known. HCV infection is strictly linked to lipoprotein metabolism, and HCV virions may initially interact with SR-BI through associated lipoproteins before subsequent direct interactions of the viral glycoproteins with SR-BI occur. The kinetics of inhibition of cell culture-derived HCV (HCVcc) infection with an anti-SR-BI monoclonal antibody imply that the recognition of SR-BI by HCV is an early event of the infection process. Swapping and single-substitution mutants between mouse and human SR-BI sequences showed reduced binding to the recombinant soluble E2 (sE2) envelope glycoprotein, thus suggesting that the SR-BI interaction with the HCV envelope is likely to involve species-specific protein elements. Most importantly, SR-BI mutants defective for sE2 binding, although retaining wild-type activity for receptor oligomerization and binding to the physiological ligand HDL, were impaired in their ability to fully restore HCVcc infectivity when transduced into an SR-BI-knocked-down Huh-7.5 cell line. These findings suggest a specific and direct role for the identified residues in binding HCV and mediating virus entry. Moreover, the observation that different regions of SR-BI are involved in HCV and HDL binding supports the hypothesis that new therapeutic strategies aimed at interfering with virus/SR-BI recognition are feasible.


Subject(s)
Hepacivirus/physiology , Receptors, Virus , Scavenger Receptors, Class B/physiology , Virus Internalization , Animals , Antibodies, Monoclonal , Cells, Cultured , Hepacivirus/immunology , Hepatitis C/immunology , Humans , Kinetics , Lipoproteins, HDL/metabolism , Mice , Scavenger Receptors, Class B/metabolism , Species Specificity
12.
J Virol ; 83(18): 9079-93, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19587042

ABSTRACT

Naturally occurring hepatitis C virus (HCV) subgenomic RNAs have been found in several HCV patients. These subgenomic deletion mutants, mostly lacking the genes encoding envelope glycoproteins, were found in both liver and serum, where their relatively high abundance suggests that they are capable of autonomous replication and can be packaged and secreted in viral particles, presumably harboring the envelope proteins from wild type virus coinfecting the same cell. We recapitulated some of these natural subgenomic deletions in the context of the isolate JFH-1 and confirmed these hypotheses in vitro. In Huh-7.5 cells, these deletion-containing genomes show robust replication and can be efficiently trans-packaged and infect naïve Huh-7.5 cells when cotransfected with the full-length wild-type J6/JFH genome. The genome structure of these natural subgenomic deletion mutants was dissected, and the maintenance of both core and NS2 regions was proven to be significant for efficient replication and trans-packaging. To further explore the requirements needed to achieve trans-complementation, we provided different combinations of structural proteins in trans. Optimal trans-complementation was obtained when fragments of the polyprotein encompassing core to p7 or E1 to NS2 were expressed. Finally, we generated a stable helper cell line, constitutively expressing the structural proteins from core to p7, which efficiently supports trans-complementation of a subgenomic deletion encompassing amino acids 284 to 732. This cell line can produce and be infected by defective particles, thus representing a powerful tool to investigate the life cycle and relevance of natural HCV subgenomic deletion mutants in vivo.


Subject(s)
Hepacivirus/genetics , Sequence Deletion , Virion , Virus Assembly , Cell Line , Genome, Viral , Humans , RNA, Viral , Viral Structural Proteins/genetics , Virus Replication
13.
J Mol Biol ; 390(5): 1048-59, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19505479

ABSTRACT

Hepatitis C virus (HCV) exists in six major genotypes. Compared with the 1b enzyme, genotype 2b HCV polymerase exhibits a more than 100-fold reduction in sensitivity to the indole-N-acetamide class of non-nucleoside inhibitors. These compounds have been shown to bind in a pocket occupied by helix A of the mobile Lambda1 loop in the apoenzyme. The three-dimensional structure of the HCV polymerase from genotype 2b was determined to 1.9-A resolution and compared with the genotype 1b enzyme. This structural analysis suggests that genotypic variants result in a different shape of the inhibitor binding site. Mutants of the inhibitor binding pocket were generated in a 1b enzyme and evaluated for their binding affinity and sensitivity to inhibition by indole-N-acetamides. Most of the point mutants showed little variation in activity and IC(50), with the exception of 15- and 7-fold increases in IC(50) for Leu392Ile and Val494Ala mutants (1b-->2b), respectively. Furthermore, a 1b replicon with 20-fold resistance to this class of inhibitors was selected and shown to contain the Leu392Ile mutation. Chimeric enzymes, where the 2b fingertip Lambda1 loop, pocket or both replaced the corresponding regions of the 1b enzyme, were also generated. The fingertip chimera retained 1b-like inhibitor binding affinity, whereas the other two chimeric constructs and the 2b enzyme displayed between 50- and 100-fold reduction in binding affinity. Together, these data suggest that differences in the amino acid composition and shape of the indole-N-acetamide binding pocket are responsible for the resistance of the 2b polymerase to this class of inhibitors.


Subject(s)
Antiviral Agents/pharmacology , DNA-Directed RNA Polymerases/chemistry , Drug Resistance, Viral , Hepacivirus/enzymology , Nucleosides/pharmacology , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Amino Acid Substitution/drug effects , Antiviral Agents/chemistry , Binding Sites , Crystallography, X-Ray , DNA-Directed RNA Polymerases/antagonists & inhibitors , DNA-Directed RNA Polymerases/isolation & purification , Drug Resistance, Viral/drug effects , Genotype , Hepacivirus/drug effects , Hepacivirus/genetics , Indoleacetic Acids/chemistry , Indoleacetic Acids/pharmacology , Kinetics , Models, Molecular , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Protein Structure, Secondary , Replicon/genetics , Structural Homology, Protein , Viral Nonstructural Proteins/isolation & purification
14.
J Virol ; 81(15): 8063-71, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17507483

ABSTRACT

The human scavenger class B type 1 receptor (SR-B1/Cla1) was identified as a putative receptor for hepatitis C virus (HCV) because it binds to soluble recombinant HCV envelope glycoprotein E2 (sE2). High-density lipoprotein (HDL), a natural SR-B1 ligand, was shown to increase the in vitro infectivity of retroviral pseudoparticles bearing HCV envelope glycoproteins and of cell culture-derived HCV (HCVcc), suggesting that SR-B1 promotes viral entry in an HDL-dependent manner. To determine whether SR-B1 participates directly in HCV infection or facilitates HCV entry through lipoprotein uptake, we generated a panel of monoclonal antibodies (MAbs) against native human SR-B1. Two of them, 3D5 and C167, bound to conformation-dependent SR-B1 determinants and inhibited the interaction of sE2 with SR-B1. These antibodies efficiently blocked HCVcc infection of Huh-7.5 hepatoma cells in a dose-dependent manner. To examine the role of HDL in SR-B1-mediated HCVcc infection, we set up conditions for HCVcc production and infection in serum-free medium. HCVcc efficiently infected Huh-7.5 cells in the absence of serum lipoproteins, and addition of HDL led to a twofold increase in infectivity. However, the HDL-induced enhancement of infection had no impact on the neutralization potency of MAb C167, despite its ability to inhibit both HDL binding to cells and SR-B1-mediated lipid transfer. Of note, MAb C167 also potently blocked Huh-7.5 infection by an HCV strain recovered from HCVcc-infected chimpanzees. These results demonstrate that SR-B1 is essential for infection with HCV produced in vitro and in vivo and suggest the possible use of anti-SR-B1 antibodies as therapeutic agents.


Subject(s)
Antibodies, Monoclonal/immunology , Hepacivirus/immunology , Hepatitis C , Lipoproteins, HDL/metabolism , Scavenger Receptors, Class B/immunology , Antibody Affinity , Cell Line , Cholesterol/metabolism , Hepatitis C/immunology , Hepatitis C/prevention & control , Humans , Scavenger Receptors, Class B/genetics
15.
Antivir Chem Chemother ; 16(4): 225-45, 2005.
Article in English | MEDLINE | ID: mdl-16130521

ABSTRACT

The high prevalence of the disease caused by hepatitis C virus (HCV) and the limited efficacy of interferon-based therapies have stimulated the search for safer and more effective drugs. The development of inhibitors of the HCV NS5B RNA polymerase represents a promising strategy for identifying novel anti-HCV therapeutics. However, the high genetic diversity, mutation rate and turnover of HCV are expected to favour the emergence of drug resistance, limiting the clinical usefulness of polymerase inhibitors. Thus, the characterization of the drug-resistance profile of these antiviral agents is considered crucial for identifying the inhibitors with a higher probability of clinical success. In the absence of an efficient in vitro infection system, HCV sub-genomic replicons have been used to study viral resistance to both nucleoside and non-nucleoside NS5B inhibitors. While these studies suggest that drug-resistant viruses are likely to evolve in vivo, they provide a wealth of information that should help in the identification of inhibitors with improved and distinct resistance profiles that might be used for combination therapy.


Subject(s)
Antiviral Agents/pharmacology , Drug Resistance, Viral , Hepacivirus/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Animals , Antiviral Agents/chemistry , Genome, Viral , Hepacivirus/genetics , Humans , Molecular Structure , Mutation , RNA-Dependent RNA Polymerase/chemistry , Viral Nonstructural Proteins/chemistry
16.
J Med Chem ; 48(14): 4547-57, 2005 Jul 14.
Article in English | MEDLINE | ID: mdl-15999993

ABSTRACT

Infections caused by hepatitis C virus (HCV) are a significant world health problem for which novel therapies are in urgent demand. Compounds that block replication of subgenomic HCV RNA in liver cells are of interest because of their demonstrated antiviral effect in the clinic. In followup to our recent report that indole-N-acetamides (e.g., 1) are potent allosteric inhibitors of the HCV NS5B polymerase enzyme, we describe here their optimization as cell-based inhibitors. The crystal structure of 1 bound to NS5B was a guide in the design of a two-dimensional compound array that highlighted that formally zwitterionic inhibitors have strong intracellular potency and that pregnane X receptor (PXR) activation (an undesired off-target activity) is linked to a structural feature of the inhibitor. Optimized analogues devoid of PXR activation (e.g., 55, EC(50) = 127 nM) retain strong cell-based efficacy under high serum conditions and show acceptable pharmacokinetics parameters in rat and dog.


Subject(s)
Acetamides/chemical synthesis , Antiviral Agents/chemical synthesis , Hepacivirus/enzymology , Indoles/chemical synthesis , RNA, Viral/drug effects , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Acetamides/chemistry , Acetamides/pharmacology , Allosteric Regulation , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Biological Availability , Cell Line, Tumor , Dogs , Genome, Viral , Half-Life , Hepacivirus/genetics , Humans , Indoles/chemistry , Indoles/pharmacology , Models, Molecular , Pregnane X Receptor , RNA-Dependent RNA Polymerase/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Steroid/agonists , Structure-Activity Relationship , Tissue Distribution , Viral Nonstructural Proteins/chemistry
17.
Biochem Biophys Res Commun ; 326(1): 66-73, 2005 Jan 07.
Article in English | MEDLINE | ID: mdl-15567153

ABSTRACT

It is well established that certain subpopulations of human adult stem cells can generate hepatocyte-like cells when transplanted into adult immunosuppressed mice. In the present study, we wanted to explore whether xeno-transplantation of human cord blood CD34(+) (hCBCD34(+)) cells during pre-immune stages of development in immunocompetent mice might also lead to human-mouse liver chimerism. Freshly isolated hCBCD34(+) cells were xeno-transplanted into non-immunosuppressed mice by both intra-blastocyst and intra-fetal injections. One and four weeks after birth, immunostaining for different human-specific hepatocyte markers: human hepatocyte-specific antigen, human serum albumin, and human alpha-1-antitrypsin indicated the presence of human hepatocyte-like cells in the livers of transplanted animals. Detection of human albumin mRNA further corroborated the development of pre-immune human-mouse chimeras. The current report, besides providing new evidence of the potential of hCBCD34(+) cells to generate human hepatocyte-like cells, suggests novel strategies for generating immunocompetent mice harboring humanized liver.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Hepatocytes/cytology , Hepatocytes/physiology , Liver/embryology , Stem Cells/cytology , Transplantation, Heterologous/methods , Animals , Cell Differentiation/physiology , Cell Proliferation , Female , Humans , Immunocompetence/physiology , Injections/methods , Liver/cytology , Liver/physiology , Mice , Mice, SCID , Pregnancy
18.
J Virol ; 78(23): 12809-16, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15542633

ABSTRACT

Hepatitis C virus (HCV) has evolved complex strategies to evade host immune responses and establish chronic infection. The only treatment available for HCV infections, alpha interferon (IFN-alpha), is effective in a limited percentage of patients. The mechanisms by which IFN-alpha interferes with the HCV life cycle and the reasons for limited effectiveness of IFN-alpha therapy have not yet been fully elucidated. Using a cell-based HCV replication system and specific kinase inhibitors, we examined the role played by various signaling pathways in the IFN-alpha-mediated HCV clearance. We reported that conventional protein kinase C (cPKC) activity is important for the effectiveness of IFN-alpha treatment. In cells treated with a cPKC-specific inhibitor, IFN-alpha failed to induce an efficient HCV RNA degradation. The lack of cPKC activity leads to a broad reduction of IFN-alpha-stimulated gene expression due to a significant impairment of STAT1 and STAT3 tyrosine phosphorylation. Thus, modulation of cPKC function by either host or viral factors could influence the positive outcome of IFN-alpha-mediated antiviral therapies.


Subject(s)
Antiviral Agents/pharmacology , DNA-Binding Proteins/metabolism , Hepacivirus/drug effects , Interferon-alpha/pharmacology , Protein Kinase C/physiology , Replicon/drug effects , Trans-Activators/metabolism , Cell Line , Gene Expression Regulation, Viral/drug effects , Hepacivirus/genetics , Humans , Protein Kinase C/antagonists & inhibitors , RNA, Viral/metabolism , STAT1 Transcription Factor , STAT3 Transcription Factor
19.
J Gen Virol ; 85(Pt 7): 1867-1875, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15218171

ABSTRACT

An efficient model is currently used to study hepatitis C virus (HCV) replication in cell culture. It involves transfection in Huh7, a hepatoma-derived cell line, of an antibiotic (neomycin) selectable HCV subgenomic replicon encoding the non-structural (NS) proteins from NS3 to NS5B. However, strong and sustained replication is achieved only on the appearance of adaptive mutations in viral proteins. The most effective of these adaptive mutations are concentrated mainly in NS5A, not only into the original Con1 but also in the recently established HCV-BK and HCV-H77 isolate-derived replicons. This suggests that the expression of wild-type (wt) NS5A may not allow efficient HCV RNA replication in cell culture. With the use of a beta-lactamase reporter gene as a marker for HCV replication and TaqMan RNA analysis, the replication of different HCV replicons in cotransfection experiments was investigated. Comparing wt with NS5A-adapted replicons, the strong evidence accumulated showed that the expression of wt NS5A was actually able to inhibit the replication of NS5A-adapted replicons. This feature was characterized as a dominant negative effect. Interestingly, an NS5B (R2884G)-adapted replicon, containing a wt NS5A, was dominant negative on an NS5A-adapted replicon but was not inhibited by the original Con1 replicon. In conclusion, these studies revealed that the original wt Con1 replicon is not only incompetent for replication in cell culture, but is also able to interfere with NS5A-adapted replicons.


Subject(s)
Hepacivirus/genetics , RNA, Viral/genetics , Replicon/genetics , Viral Nonstructural Proteins/genetics , Amino Acid Substitution , Carcinoma, Hepatocellular , Cell Line, Tumor , Genes, Reporter , Genome, Viral , Hepacivirus/physiology , Humans , Liver Neoplasms , Mutagenesis, Site-Directed , Transfection , Virus Replication/genetics
20.
J Virol ; 77(6): 3669-79, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12610142

ABSTRACT

The hepatitis C virus (HCV) serine protease is necessary for viral replication and represents a valid target for developing new therapies for HCV infection. Potent and selective inhibitors of this enzyme have been identified and shown to inhibit HCV replication in tissue culture. The optimization of these inhibitors for clinical development would greatly benefit from in vitro systems for the identification and the study of resistant variants. We report the use HCV subgenomic replicons to isolate and characterize mutants resistant to a protease inhibitor. Taking advantage of the replicons' ability to transduce resistance to neomycin, we selected replicons with decreased sensitivity to the inhibitor by culturing the host cells in the presence of the inhibitor and neomycin. The selected replicons replicated to the same extent as those in parental cells. Sequence analysis followed by transfection of replicons containing isolated mutations revealed that resistance was mediated by amino acid substitutions in the protease. These results were confirmed by in vitro experiments with mutant enzymes and by modeling the inhibitor in the three-dimensional structure of the protease.


Subject(s)
Carrier Proteins/genetics , Drug Resistance, Viral/genetics , Genetic Variation , Hepacivirus/drug effects , Serine Proteinase Inhibitors/pharmacology , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics , Binding Sites/drug effects , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/drug effects , Hepacivirus/enzymology , Hepacivirus/genetics , Humans , Intracellular Signaling Peptides and Proteins , Microbial Sensitivity Tests , Models, Molecular , Mutation , Replicon , Selection, Genetic , Tumor Cells, Cultured , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/drug effects , Viral Proteins/antagonists & inhibitors , Viral Proteins/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...