Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Prod Res ; : 1-10, 2023 Dec 09.
Article in English | MEDLINE | ID: mdl-38069761

ABSTRACT

The phytochemical study of Cattleya intermedia (Orchidaceae) led to the isolation of two new stilbenoids and one new 9,10-dihydrophenanthrene, 4',5-dihydroxy-2',3-dimethoxy-dihydrostilbene (1), 3,6'-dihydroxy-4'-methoxy-dihydrostilbene (2) and 1,2,6-trihydroxy-3,8-dimethoxy-9,10-dihydrophenanthrene (3), named cattleymediol, cattleyol and phenanmediol, respectively, in addition to other five known compounds (4-8). The structural elucidations of the isolated compounds were carried out through the analyses of the one-dimensional 1H,1³C and NOE NMR spectra, and the 2D HSQC, HMBC, COSY and NOESY spectra, besides high-resolution mass spectrometry. In addition to this, the crude extract and its main fractions were analysed by ultra-high performance liquid chromatography coupled to high resolution mass spectrometry (UHPLC-QTOF-MS/MS), leading to the putative identification of several other compounds, including flavonoids and organic acids derivatives. Finally, the main fractions of the crude extract, and the pure compounds cattleymediol (1) and lusiantridine (7), had their antiproliferative activities evaluated against human cancerous HeLa and non-cancerous VERO cells.

2.
J Hepatol ; 70(6): 1082-1092, 2019 06.
Article in English | MEDLINE | ID: mdl-30769006

ABSTRACT

BACKGROUND & AIMS: Hepatitis C virus (HCV) infection causes chronic liver disease. Antivirals have been developed and cure infection. However, resistance can emerge and salvage therapies with alternative modes of action could be useful. Several licensed drugs have emerged as HCV entry inhibitors and are thus candidates for drug repurposing. We aimed to dissect their mode of action, identify improved derivatives and determine their viral targets. METHODS: HCV entry inhibition was tested for a panel of structurally related compounds, using chimeric viruses representing diverse genotypes, in addition to viruses containing previously determined resistance mutations. Chemical modeling and synthesis identified improved derivatives, while generation of susceptible and non-susceptible chimeric viruses pinpointed E1 determinants of compound sensitivity. RESULTS: Molecules of the diphenylpiperazine, diphenylpiperidine, phenothiazine, thioxanthene, and cycloheptenepiperidine chemotypes inhibit HCV infection by interfering with membrane fusion. These molecules and a novel p-methoxy-flunarizine derivative with improved efficacy preferentially inhibit genotype 2 viral strains. Viral residues within a central hydrophobic region of E1 (residues 290-312) control susceptibility. At the same time, viral features in this region also govern pH-dependence of viral membrane fusion. CONCLUSIONS: Small molecules from different chemotypes related to flunarizine preferentially inhibit HCV genotype 2 membrane fusion. A hydrophobic region proximal to the putative fusion loop controls sensitivity to these drugs and the pH range of membrane fusion. An algorithm considering viral features in this region predicts viral sensitivity to membrane fusion inhibitors. Resistance to flunarizine correlates with more relaxed pH requirements for fusion. LAY SUMMARY: This study describes diverse compounds that act as HCV membrane fusion inhibitors. It defines viral properties that determine sensitivity to these molecules and thus provides information to identify patients that may benefit from treatment with membrane fusion inhibitors.


Subject(s)
Hepacivirus/drug effects , Virus Internalization/drug effects , Antiviral Agents/pharmacology , Drug Resistance, Viral , Flunarizine/pharmacology , Hepacivirus/physiology , Humans , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Structure-Activity Relationship
3.
Nat Prod Res ; 32(24): 2916-2921, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29117727

ABSTRACT

The phytochemical study of Laelia marginata (Lindl.) L. O. Williams (Orchidaceae) led to the isolation of a new natural product named crispoic acid (1), together with six other known compounds (2-7). The new natural product was identified as a dimer of eucomic acid and was structurally characterised based upon 1D and 2D NMR and HRMS data. Biological assays with plant crude extract, fractions and isolated compounds were performed against two human cancer cell lines (Hela and Siha), and the tropical parasites Trypanosoma cruzi and Leishmania (Leishmania) amazonensis. The phenantrenoid 9,10-dihydro-4-methoxyphenanthren-2,7-diol 2 was active against Hela and Siha cells (CC50 5.86 ± 0.19 and 20.78 ± 2.72 µg/mL, respectively). Sub-lethal concentrations of the flavone rhamnazin 4 were not able to rescue the viability of the Vero cells infected by Zika virus.


Subject(s)
Antineoplastic Agents, Phytogenic/isolation & purification , Antiparasitic Agents/isolation & purification , Chlorocebus aethiops , Orchidaceae/chemistry , Parasites/drug effects , Zika Virus/drug effects , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antiparasitic Agents/pharmacology , Cell Line , Cell Line, Tumor , Humans , Leishmania/drug effects , Molecular Structure , Phytochemicals/isolation & purification , Phytochemicals/pharmacology , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Trypanosoma cruzi/drug effects , Vero Cells/virology
4.
Antiviral Res ; 147: 19-28, 2017 11.
Article in English | MEDLINE | ID: mdl-28923507

ABSTRACT

Approximately 142 million people worldwide are infected with hepatitis C virus (HCV). Although potent direct acting antivirals are available, high costs limit access to treatment. Chronic hepatitis C virus infection remains a major cause of orthotopic liver transplantation. Moreover, re-infection of the graft occurs regularly. Antivirals derived from natural sources might be an alternative and cost-effective option to complement therapy regimens for global control of hepatitis C virus infection. We tested the antiviral properties of a mixture of different Chinese herbs/roots named Zhi Bai Di Huang Wan (ZBDHW) and its individual components on HCV. One of the ZBDHW components, Penta-O-Galloyl-Glucose (PGG), was further analyzed for its mode of action in vitro, its antiviral activity in primary human hepatocytes as well as for its bioavailability and hepatotoxicity in mice. ZBDHW, its component Cortex Moutan and the compound PGG efficiently block entry of HCV of all major genotypes and also of the related flavivirus Zika virus. PGG does not disrupt HCV virion integrity and acts primarily during virus attachment. PGG shows an additive effect when combined with the well characterized HCV inhibitor Daclatasvir. Analysis of bioavailability in mice revealed plasma levels above tissue culture IC50 after a single intraperitoneal injection. In conclusion, PGG is a pangenotypic HCV entry inhibitor with high bioavailability. The low cost and wide availability of this compound make it a promising candidate for HCV combination therapies, and also emerging human pathogenic flaviviruses like ZIKV.


Subject(s)
Antiviral Agents/pharmacology , Drugs, Chinese Herbal/chemistry , Hepacivirus/drug effects , Hydrolyzable Tannins/pharmacology , Paeonia/chemistry , Virus Attachment/drug effects , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Biological Availability , Carbamates , Cell Line, Tumor , Cells, Cultured , Drug Synergism , Drugs, Chinese Herbal/pharmacology , Hepacivirus/genetics , Hepacivirus/physiology , Hepatitis C/drug therapy , Hepatitis C, Chronic/drug therapy , Hepatocytes/drug effects , Humans , Hydrolyzable Tannins/administration & dosage , Hydrolyzable Tannins/pharmacokinetics , Imidazoles/pharmacology , Mice , Mice, SCID , Plant Extracts/chemistry , Plant Extracts/pharmacology , Pyrrolidines , Valine/analogs & derivatives , Virion/drug effects , Virus Replication/drug effects
5.
Hepatology ; 63(1): 49-62, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26248546

ABSTRACT

UNLABELLED: To explore mechanisms of hepatitis C viral (HCV) replication we screened a compound library including licensed drugs. Flunarizine, a diphenylmethylpiperazine used to treat migraine, inhibited HCV cell entry in vitro and in vivo in a genotype-dependent fashion. Analysis of mosaic viruses between susceptible and resistant strains revealed that E1 and E2 glycoproteins confer susceptibility to flunarizine. Time of addition experiments and single particle tracking of HCV demonstrated that flunarizine specifically prevents membrane fusion. Related phenothiazines and pimozide also inhibited HCV infection and preferentially targeted HCV genotype 2 viruses. However, phenothiazines and pimozide exhibited improved genotype coverage including the difficult to treat genotype 3. Flunarizine-resistant HCV carried mutations within the alleged fusion peptide and displayed cross-resistance to these compounds, indicating that these drugs have a common mode of action. CONCLUSION: These observations reveal novel details about HCV membrane fusion; moreover, flunarizine and related compounds represent first-in-class HCV fusion inhibitors that merit consideration for repurposing as a cost-effective component of HCV combination therapies.


Subject(s)
Flunarizine/pharmacology , Hepacivirus/drug effects , Viral Fusion Proteins/drug effects , Virus Internalization/drug effects , Cells, Cultured , Genotype , Hepacivirus/genetics , Humans , Viral Fusion Proteins/genetics
6.
Cell Rep ; 12(5): 864-78, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26212323

ABSTRACT

Hepatitis C virus (HCV) enters human hepatocytes through a multistep mechanism involving, among other host proteins, the virus receptor CD81. How CD81 governs HCV entry is poorly characterized, and CD81 protein interactions after virus binding remain elusive. We have developed a quantitative proteomics protocol to identify HCV-triggered CD81 interactions and found 26 dynamic binding partners. At least six of these proteins promote HCV infection, as indicated by RNAi. We further characterized serum response factor binding protein 1 (SRFBP1), which is recruited to CD81 during HCV uptake and supports HCV infection in hepatoma cells and primary human hepatocytes. SRFBP1 facilitates host cell penetration by all seven HCV genotypes, but not of vesicular stomatitis virus and human coronavirus. Thus, SRFBP1 is an HCV-specific, pan-genotypic host entry factor. These results demonstrate the use of quantitative proteomics to elucidate pathogen entry and underscore the importance of host protein-protein interactions during HCV invasion.


Subject(s)
Hepacivirus/physiology , Proteomics , Transcription Factors/metabolism , Virus Internalization , Cell Line, Tumor , Humans
7.
J Hepatol ; 63(4): 813-21, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26070407

ABSTRACT

BACKGROUND & AIMS: Soraphen A (SorA) is a myxobacterial metabolite that inhibits the acetyl-CoA carboxylase, a key enzyme in lipid biosynthesis. We have previously identified SorA to efficiently inhibit the human immunodeficiency virus (HIV). The aim of the present study was to evaluate the capacity of SorA and analogues to inhibit hepatitis C virus (HCV) infection. METHODS: SorA inhibition capacity was evaluated in vitro using cell culture derived HCV, HCV pseudoparticles and subgenomic replicons. Infection studies were performed in the hepatoma cell line HuH7/Scr and in primary human hepatocytes. The effects of SorA on membranous web formation were analysed by electron microscopy. RESULTS: SorA potently inhibits HCV infection at nanomolar concentrations. Obtained EC50 values were 0.70 nM with a HCV reporter genome, 2.30 nM with wild-type HCV and 2.52 nM with subgenomic HCV replicons. SorA neither inhibited HCV RNA translation nor HCV entry, as demonstrated with subgenomic HCV replicons and HCV pseudoparticles, suggesting an effect on HCV replication. Consistent with this, evidence was obtained that SorA interferes with formation of the membranous web, the site of HCV replication. Finally, a series of natural and synthetic SorA analogues helped to establish a first structure-activity relationship. CONCLUSIONS: SorA has a very potent anti-HCV activity. Since it also interferes with the membranous web formation, SorA is an excellent tool to unravel the mechanism of HCV replication.


Subject(s)
Hepacivirus/genetics , Hepatitis C/drug therapy , Hepatocytes/drug effects , Macrolides/pharmacology , RNA, Viral/genetics , Virus Replication/drug effects , Antiviral Agents/pharmacology , Cell Line , Hepacivirus/drug effects , Hepatitis C/pathology , Hepatitis C/virology , Hepatocytes/ultrastructure , Hepatocytes/virology , Humans , Microscopy, Electron
8.
Hepatology ; 62(3): 702-14, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25999047

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) is a positive-strand RNA virus that primarily infects human hepatocytes. Infections with HCV constitute a global health problem, with 180 million people currently chronically infected. Recent studies have reported that cholesterol 25-hydroxylase (CH25H) is expressed as an interferon-stimulated gene and mediates antiviral activities against different enveloped viruses through the production of 25-hydroxycholesterol (25HC). However, the intrinsic regulation of human CH25H (hCH25H) expression within the liver as well as its mechanistic effects on HCV infectivity remain elusive. In this study, we characterized the expression of hCH25H using liver biopsies and primary human hepatocytes. In addition, the antiviral properties of this protein and its enzymatic product, 25HC, were further characterized against HCV in tissue culture. Levels of hCH25H messenger RNA were significantly up-regulated both in HCV-positive liver biopsies and in HCV-infected primary human hepatocytes. The expression of hCH25H in primary human hepatocytes was primarily and transiently induced by type I interferon. Transient expression of hCH25H in human hepatoma cells restricted HCV infection in a genotype-independent manner. This inhibition required the enzymatic activity of CH25H. We observed an inhibition of viral membrane fusion during the entry process by 25HC, which was not due to a virucidal effect. Yet the primary effect by 25HC on HCV was at the level of RNA replication, which was observed using subgenomic replicons of two different genotypes. Further analysis using electron microscopy revealed that 25HC inhibited formation of the membranous web, the HCV replication factory, independent of RNA replication. CONCLUSION: Infection with HCV causes up-regulation of interferon-inducible CH25H in vivo, and its product, 25HC, restricts HCV primarily at the level of RNA replication by preventing formation of the viral replication factory.


Subject(s)
Hepacivirus/genetics , Interferons/pharmacology , Steroid Hydroxylases/genetics , Virus Internalization/drug effects , Virus Replication/drug effects , Biopsy, Needle , Cells, Cultured , DNA Replication/drug effects , Gene Expression Regulation, Viral , Hepatitis C, Chronic/pathology , Hepatocytes/metabolism , Humans , Sensitivity and Specificity , Up-Regulation/drug effects
9.
PLoS Pathog ; 9(5): e1003355, 2013.
Article in English | MEDLINE | ID: mdl-23658526

ABSTRACT

Hepatitis C virus (HCV) p7 is a membrane-associated ion channel protein crucial for virus production. To analyze how p7 contributes to this process, we dissected HCV morphogenesis into sub-steps including recruitment of HCV core to lipid droplets (LD), virus capsid assembly, unloading of core protein from LDs and subsequent membrane envelopment of capsids. Interestingly, we observed accumulation of slowly sedimenting capsid-like structures lacking the viral envelope in cells transfected with HCV p7 mutant genomes which possess a defect in virion production. Concomitantly, core protein was enriched at the surface of LDs. This indicates a defect in core/capsid unloading from LDs and subsequent membrane envelopment rather than defective trafficking of core to this cellular organelle. Protease and ribonuclease digestion protection assays, rate zonal centrifugation and native, two dimensional gel electrophoresis revealed increased amounts of high-order, non-enveloped core protein complexes unable to protect viral RNA in cells transfected with p7 mutant genomes. These results suggest accumulation of capsid assembly intermediates that had not yet completely incorporated viral RNA in the absence of functional p7. Thus, functional p7 is necessary for the final steps of capsid assembly as well as for capsid envelopment. These results support a model where capsid assembly is linked with membrane envelopment of nascent RNA-containing core protein multimers, a process coordinated by p7. In summary, we provide novel insights into the sequence of HCV assembly events and essential functions of p7.


Subject(s)
Capsid/metabolism , Hepacivirus/physiology , Hepatitis C/metabolism , RNA, Viral/metabolism , Viral Proteins/metabolism , Virus Assembly/physiology , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/virology , Hepacivirus/ultrastructure , Hepatitis C/genetics , Hepatitis C/pathology , Humans , RNA, Viral/genetics , Viral Proteins/genetics
10.
PLoS One ; 7(4): e35876, 2012.
Article in English | MEDLINE | ID: mdl-22558251

ABSTRACT

The type II transmembrane serine proteases TMPRSS2 and HAT activate influenza viruses and the SARS-coronavirus (TMPRSS2) in cell culture and may play an important role in viral spread and pathogenesis in the infected host. However, it is at present largely unclear to what extent these proteases are expressed in viral target cells in human tissues. Here, we show that both HAT and TMPRSS2 are coexpressed with 2,6-linked sialic acids, the major receptor determinant of human influenza viruses, throughout the human respiratory tract. Similarly, coexpression of ACE2, the SARS-coronavirus receptor, and TMPRSS2 was frequently found in the upper and lower aerodigestive tract, with the exception of the vocal folds, epiglottis and trachea. Finally, activation of influenza virus was conserved between human, avian and porcine TMPRSS2, suggesting that this protease might activate influenza virus in reservoir-, intermediate- and human hosts. In sum, our results show that TMPRSS2 and HAT are expressed by important influenza and SARS-coronavirus target cells and could thus support viral spread in the human host.


Subject(s)
Disease Reservoirs/veterinary , Gastrointestinal Tract/enzymology , Influenza, Human/enzymology , Respiratory System/enzymology , Serine Endopeptidases/genetics , Severe Acute Respiratory Syndrome/enzymology , Angiotensin-Converting Enzyme 2 , Animals , Birds , Cell Line , Disease Reservoirs/virology , Enzyme Activation , Gastrointestinal Tract/virology , Gene Expression , Humans , Influenza, Human/genetics , Influenza, Human/transmission , Influenza, Human/virology , Orthomyxoviridae/physiology , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Respiratory System/virology , Severe acute respiratory syndrome-related coronavirus/physiology , Serine Endopeptidases/metabolism , Severe Acute Respiratory Syndrome/genetics , Severe Acute Respiratory Syndrome/transmission , Severe Acute Respiratory Syndrome/virology , Sialic Acids/metabolism , Swine
11.
Int J Mol Sci ; 12(9): 5815-27, 2011.
Article in English | MEDLINE | ID: mdl-22016628

ABSTRACT

AIMS: The relationship between variants in SLCO1B1 and SLCO2B1 genes and lipid-lowering response to atorvastatin was investigated. MATERIAL AND METHODS: One-hundred-thirty-six unrelated individuals with hypercholesterolemia were selected and treated with atorvastatin (10 mg/day/4 weeks). They were genotyped with a panel of ancestry informative markers for individual African component of ancestry (ACA) estimation by SNaPshot(®) and SLCO1B1 (c.388A>G, c.463C>A and c.521T>C) and SLCO2B1 (-71T>C) gene polymorphisms were identified by TaqMan(®) Real-time PCR. RESULTS: Subjects carrying SLCO1B1 c.388GG genotype exhibited significantly high low-density lipoprotein (LDL) cholesterol reduction relative to c.388AA+c.388AG carriers (41 vs. 37%, p = 0.034). Haplotype analysis revealed that homozygous of SLCO1B1*15 (c.521C and c.388G) variant had similar response to statin relative to heterozygous and non-carriers. A multivariate logistic regression analysis confirmed that c.388GG genotype was associated with higher LDL cholesterol reduction in the study population (OR: 3.2, CI95%:1.3-8.0, p < 0.05). CONCLUSION: SLCO1B1 c.388A>G polymorphism causes significant increase in atorvastatin response and may be an important marker for predicting efficacy of lipid-lowering therapy.


Subject(s)
Heptanoic Acids/therapeutic use , Hypercholesterolemia/drug therapy , Hypercholesterolemia/genetics , Organic Anion Transporters/genetics , Polymorphism, Single Nucleotide , Pyrroles/therapeutic use , Aged , Anticholesteremic Agents/therapeutic use , Atorvastatin , Female , Gene Frequency , Genotype , Haplotypes , Humans , Linkage Disequilibrium , Liver-Specific Organic Anion Transporter 1 , Logistic Models , Male , Middle Aged , Multivariate Analysis , Pharmacogenetics/methods , Treatment Outcome
12.
Int J Mol Sci ; 12(9): 5815-5827, 2011. ilus, tab
Article in English | Sec. Est. Saúde SP, SESSP-IDPCPROD, Sec. Est. Saúde SP | ID: biblio-1063493

ABSTRACT

Aims: The relationship between variants in SLCO1B1 and SLCO2B1 genes and lipid-lowering response to atorvastatin was investigated. Material and Methods: One-hundred-thirty-six unrelated individuals with hypercholesterolemia were selected andOPEN ACCESStreated with atorvastatin (10 mg/day/4 weeks). They were genotyped with a panel of ancestry informative markers for individual African component of ancestry (ACA) estimation by SNaPshot® and SLCO1B1 (c.388A>G, c.463C>A and c.521T>C) and SLCO2B1 (−71T>C) gene polymorphisms were identified by TaqMan® Real-time PCR. Results: Subjects carrying SLCO1B1 c.388GG genotype exhibited significantly high low-density lipoprotein (LDL) cholesterol reduction relative to c.388AA+c.388AG carriers (41 vs. 37%, p = 0.034). Haplotype analysis revealed that homozygous of SLCO1B1*15 (c.521C and c.388G) variant had similar response to statin relative to heterozygous and non-carriers. A multivariate logistic regression analysis confirmed that c.388GG genotype was associated with higher LDL cholesterol reduction in the study population (OR: 3.2, CI95%:1.3–8.0, p G polymorphism causes significant increase in atorvastatin response and may be an important marker for predicting efficacy of lipid-lowering therapy.


Subject(s)
Pharmacogenetics , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL
...