Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 18(8): e1010798, 2022 08.
Article in English | MEDLINE | ID: mdl-36007070

ABSTRACT

Hepatitis E virus (HEV) infection is the most common cause of acute viral hepatitis worldwide. Hepatitis E is usually asymptomatic and self-limiting but it can become chronic in immunocompromised patients and is associated with increased fulminant hepatic failure and mortality rates in pregnant women. HEV genome encodes three proteins including the ORF2 protein that is the viral capsid protein. Interestingly, HEV produces 3 isoforms of the ORF2 capsid protein which are partitioned in different subcellular compartments and perform distinct functions in the HEV lifecycle. Notably, the infectious ORF2 (ORF2i) protein is the structural component of virions, whereas the genome-free secreted and glycosylated ORF2 proteins likely act as a humoral immune decoy. Here, by using a series of ORF2 capsid protein mutants expressed in the infectious genotype 3 p6 HEV strain as well as chimeras between ORF2 and the CD4 glycoprotein, we demonstrated how an Arginine-Rich Motif (ARM) located in the ORF2 N-terminal region controls the fate and functions of ORF2 isoforms. We showed that the ARM controls ORF2 nuclear translocation likely to promote regulation of host antiviral responses. This motif also regulates the dual topology and functionality of ORF2 signal peptide, leading to the production of either cytosolic infectious ORF2i or reticular non-infectious glycosylated ORF2 forms. It serves as maturation site of glycosylated ORF2 by furin, and promotes ORF2-host cell membrane interactions. The identification of ORF2 ARM as a unique central regulator of the HEV lifecycle uncovers how viruses settle strategies to condense their genetic information and hijack cellular processes.


Subject(s)
Hepatitis E virus , Hepatitis E , Amino Acid Motifs , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Female , Glycosylation , Hepatitis E/genetics , Hepatitis E/metabolism , Hepatitis E virus/growth & development , Humans , Pregnancy
2.
PLoS Pathog ; 18(5): e1010498, 2022 05.
Article in English | MEDLINE | ID: mdl-35587469

ABSTRACT

Drug repurposing has the advantage of shortening regulatory preclinical development steps. Here, we screened a library of drug compounds, already registered in one or several geographical areas, to identify those exhibiting antiviral activity against SARS-CoV-2 with relevant potency. Of the 1,942 compounds tested, 21 exhibited a substantial antiviral activity in Vero-81 cells. Among them, clofoctol, an antibacterial drug used for the treatment of bacterial respiratory tract infections, was further investigated due to its favorable safety profile and pharmacokinetic properties. Notably, the peak concentration of clofoctol that can be achieved in human lungs is more than 20 times higher than its IC50 measured against SARS-CoV-2 in human pulmonary cells. This compound inhibits SARS-CoV-2 at a post-entry step. Lastly, therapeutic treatment of human ACE2 receptor transgenic mice decreased viral load, reduced inflammatory gene expression and lowered pulmonary pathology. Altogether, these data strongly support clofoctol as a therapeutic candidate for the treatment of COVID-19 patients.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Animals , Antiviral Agents/pharmacology , Chlorobenzenes , Chlorocebus aethiops , Cresols , Humans , Lung , Mice , Vero Cells
3.
Infect Agent Cancer ; 16(1): 69, 2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34922563

ABSTRACT

BACKGROUND: Despite recent advancements, limitations in the treatment and control of hepatitis C virus (HCV) infection reprioritized the studies for invention of an efficient HCV vaccine to elicit strong neutralizing antibodies (NAbs) and cellular responses. METHODS: Herein, we report molecular construction of a BacMam virus-based surface display for a subtype-1a HCV gpE2 (Bac-CMV-E2-gp64; Bac) that both expressed and displayed gpE2 in mammalian cells and bacouloviral envelope, respectively. RESULTS: Assessments by western blotting, Immunofluorescence and Immunogold-electron microscopy indicated the proper expression and incorporation in insect cell and baculovirus envelope, respectively. Mice immunized in three different prime-boost immunization groups of: Bac/Bac, Bac/Pro (bacoulovirus-derived gpE2) and Bac/DNA (plasmid DNA (pCDNA)-encoding gpE2) developed high levels of IgG and IFN-γ (highest for Bac/Bac group) indicating the induction of both humeral and cellular immune responses. Calculation of the IgG2a/IgG1 and IFN-γ/IL-4 ratios indicated a Th1 polarization of immune responses in the Bac/Bac and Bac/DNA groups but a balanced Th1-Th2 phenotype in the Bac/Pro group. Sera of the mice in the Bac/Bac group provided the highest percentage of cross-NAbs against a subtype-2a HCVcc (JFH1) compared to Bac/Pro and Bac/DNA groups (62% versus 41% and 6%). CONCLUSIONS: Results indicated that BacMam virus-based surface display for gpE2 might act as both subunit and DNA vaccine and offers a promising strategy for development of HCV vaccine for concurrent induction of strong humoral and cellular immune responses.

4.
Cell Mol Gastroenterol Hepatol ; 11(1): 185-198, 2021.
Article in English | MEDLINE | ID: mdl-32861832

ABSTRACT

BACKGROUND & AIMS: Infection with hepatitis C virus (HCV) remains a major cause of morbidity and mortality worldwide despite the recent advent of highly effective direct-acting antivirals. The envelope glycoproteins of HCV are heavily glycosylated with a high proportion of high-mannose glycans (HMGs), which serve as a shield against neutralizing antibodies and assist in the interaction with cell-entry receptors. However, there is no approved therapeutic targeting this potentially druggable biomarker. METHODS: The anti-HCV activity of a fusion protein consisting of Avaren lectin and the fragment crystallizable (Fc) region of a human immunoglobulin G1 antibody, Avaren-Fc (AvFc) was evaluated through the use of in vitro neutralization assays as well as an in vivo challenge in a chimeric human liver (PXB) mouse model. Drug toxicity was assessed by histopathology, serum alanine aminotransferase, and mouse body weights. RESULTS: AvFc was capable of neutralizing cell culture-derived HCV in a genotype-independent manner, with 50% inhibitory concentration values in the low nanomolar range. Systemic administration of AvFc in a histidine-based buffer was well tolerated; after 11 doses every other day at 25 mg/kg there were no significant changes in body or liver weights or in blood human albumin or serum alanine aminotransferase activity. Gross necropsy and liver pathology confirmed the lack of toxicity. This regimen successfully prevented genotype 1a HCV infection in all animals, although an AvFc mutant lacking HMG binding activity failed. CONCLUSIONS: These results suggest that targeting envelope HMGs is a promising therapeutic approach against HCV infection, and AvFc may provide a safe and efficacious means to prevent recurrent infection upon liver transplantation in HCV-related end-stage liver disease patients.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis C, Chronic/drug therapy , Immunoconjugates/pharmacology , Lectins/pharmacology , Animals , Antiviral Agents/therapeutic use , Disease Models, Animal , Female , Hepacivirus/drug effects , Hepacivirus/isolation & purification , Hepatitis C, Chronic/pathology , Hepatitis C, Chronic/virology , Hepatocytes/transplantation , Humans , Immunoconjugates/genetics , Immunoconjugates/therapeutic use , Lectins/genetics , Lectins/therapeutic use , Liver/drug effects , Liver/pathology , Liver/virology , Male , Mice , Polysaccharides/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Transplantation Chimera , Viral Envelope Proteins
5.
Sci Rep ; 8(1): 7782, 2018 May 14.
Article in English | MEDLINE | ID: mdl-29760472

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

6.
Sci Rep ; 8(1): 2092, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29391557

ABSTRACT

The human interferon (IFN)-induced MxA protein is a key antiviral host restriction factor exhibiting broad antiviral activity against many RNA viruses, including highly pathogenic avian influenza A viruses (IAV) of the H5N1 and H7N7 subtype. To date the mechanism for how MxA exerts its antiviral activity is unclear, however, additional cellular factors are believed to be essential for this activity. To identify MxA cofactors we performed a genome-wide siRNA-based screen in human airway epithelial cells (A549) constitutively expressing MxA using an H5N1 reporter virus. These data were complemented with a proteomic screen to identify MxA-interacting proteins. The combined data identified SMARCA2, the ATPase subunit of the BAF chromatin remodeling complex, as a crucial factor required for the antiviral activity of MxA against IAV. Intriguingly, our data demonstrate that although SMARCA2 is essential for expression of some IFN-stimulated genes (ISGs), and the establishment of an antiviral state, it is not required for expression of MxA, suggesting an indirect effect on MxA activity. Transcriptome analysis of SMARCA2-depleted A549-MxA cells identified a small set of SMARCA2-regulated factors required for activity of MxA, in particular IFITM2 and IGFBP3. These findings reveal that several virus-inducible factors work in concert to enable MxA restriction of IAV.


Subject(s)
Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H7N7 Subtype/growth & development , Influenza, Human/virology , Myxovirus Resistance Proteins/metabolism , Transcription Factors/metabolism , A549 Cells , Antiviral Agents/pharmacology , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Influenza A Virus, H5N1 Subtype/drug effects , Influenza A Virus, H7N7 Subtype/drug effects , Influenza, Human/drug therapy , Influenza, Human/metabolism , Interferons/pharmacology , Myxovirus Resistance Proteins/genetics , Proteome/analysis , Transcription Factors/genetics , Virus Replication
7.
J Virol ; 90(19): 8422-34, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27412600

ABSTRACT

UNLABELLED: Aminoquinolines and piperazines, linked or not, have been used successfully to treat malaria, and some molecules of this family also exhibit antiviral properties. Here we tested several derivatives of 4-aminoquinolines and piperazines for their activity against hepatitis C virus (HCV). We screened 11 molecules from three different families of compounds, and we identified anti-HCV activity in cell culture for six of them. Of these, we selected a compound (B5) that is currently ending clinical phase I evaluation for neurodegenerative diseases. In hepatoma cells, B5 inhibited HCV infection in a pangenotypic and dose-dependent manner, and its antiviral activity was confirmed in primary hepatocytes. B5 also inhibited infection by pseudoparticles expressing HCV envelope glycoproteins E1 and E2, and we demonstrated that it affects a postattachment stage of the entry step. Virus with resistance to B5 was selected by sequential passage in the presence of the drug, and reverse genetics experiments indicated that resistance was conferred mainly by a single mutation in the putative fusion peptide of E1 envelope glycoprotein (F291I). Furthermore, analyses of the effects of other closely related compounds on the B5-resistant mutant suggest that B5 shares a mode of action with other 4-aminoquinoline-based molecules. Finally, mice with humanized liver that were treated with B5 showed a delay in the kinetics of the viral infection. In conclusion, B5 is a novel interesting anti-HCV molecule that could be used to decipher the early steps of the HCV life cycle. IMPORTANCE: In the last 4 years, HCV therapy has been profoundly improved with the approval of direct-acting antivirals in clinical practice. Nevertheless, the high costs of these drugs limit access to therapy in most countries. The present study reports the identification and characterization of a compound (B5) that inhibits HCV propagation in cell culture and is currently ending clinical phase I evaluation for neurodegenerative diseases. This molecule inhibits the HCV life cycle by blocking virus entry. Interestingly, after selection of drug-resistant virus, a resistance mutation in the putative fusion peptide of E1 envelope glycoprotein was identified, indicating that B5 could be used to further investigate the fusion mechanism. Furthermore, mice with humanized liver treated with B5 showed a delay in the kinetics of the viral infection. In conclusion, B5 is a novel interesting anti-HCV molecule that could be used to decipher the early steps of the HCV life cycle.


Subject(s)
Aminoquinolines/pharmacology , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Aminoquinolines/chemistry , Aminoquinolines/isolation & purification , Animals , Antiviral Agents/chemistry , Antiviral Agents/isolation & purification , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Resistance, Viral , Hepatitis C/drug therapy , Hepatocytes/virology , Humans , Mice , Mice, SCID , Models, Molecular , Molecular Structure , Mutation, Missense , Reverse Genetics , Treatment Outcome , Viral Envelope Proteins/genetics , Virus Internalization/drug effects
8.
PLoS One ; 10(11): e0142539, 2015.
Article in English | MEDLINE | ID: mdl-26561856

ABSTRACT

The clinical course of Hepatitis C Virus (HCV) infection is highly variable between infected individual hosts: up to 80% of acutely HCV infected patients develop a chronic infection while 20% clear infection spontaneously. Spontaneous clearance of HCV infection can be predicted by several factors, including symptomatic acute infection, favorable IFNL3 polymorphisms and gender. In our study, we explored the possibility that variants in HCV cell entry factors might be involved in resistance to HCV infection. In a same case patient highly exposed but not infected by HCV, we previously identified one mutation in claudin-6 (CLDN6) and a rare variant in occludin (OCLN), two tight junction proteins involved in HCV entry into hepatocytes. Here, we conducted an extensive functional study to characterize the ability of these two natural variants to prevent HCV entry. We used lentiviral vectors to express Wildtype or mutated CLDN6 and OCLN in different cell lines and primary human hepatocytes. HCV infection was then investigated using cell culture produced HCV particles (HCVcc) as well as HCV pseudoparticles (HCVpp) expressing envelope proteins from different genotypes. Our results show that variants of CLDN6 and OCLN expressed separately or in combination did not affect HCV infection nor cell-to-cell transmission. Hence, our study highlights the complexity of HCV resistance mechanisms supporting the fact that this process probably not primarily involves HCV entry factors and that other unknown host factors may be implicated.


Subject(s)
Claudins/immunology , Hepacivirus/immunology , Hepatitis C/immunology , Occludin/immunology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Cells, Cultured , Claudins/genetics , Claudins/metabolism , Disease Resistance/genetics , Disease Resistance/immunology , Flow Cytometry , HEK293 Cells , Hep G2 Cells , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/virology , Host-Pathogen Interactions/immunology , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Microscopy, Fluorescence , Mutation/immunology , Occludin/genetics , Occludin/metabolism , Virion/immunology , Virion/physiology
9.
ACS Infect Dis ; 1(8): 380-7, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26284260

ABSTRACT

Ebola virus (EBOV), a member of the family Filoviridae, is a nonsegmented negative-sense RNA virus that causes severe, often lethal, disease in humans. EBOV RNA synthesis is carried out by a complex that includes several viral proteins. The function of this machinery is essential for viral gene expression and viral replication and is therefore a potential target for antivirals. We developed and optimized a high-throughput screening (HTS) assay based on an EBOV minigenome assay, which assesses the function of the polymerase complex. The assay is robust in 384-well format and displays a large signal to background ratio and high Z-factor values. We performed a pilot screen of 2080 bioactive compounds, identifying 31 hits (1.5% of the library) with >70% inhibition of EBOV minigenome activity. We further identified eight compounds with 50% inhibitory concentrations below their 50% cytotoxic concentrations, five of which had selectivity index (SI) values >10, suggesting specificity against the EBOV polymerase complex. These included an inhibitor of inosine monophosphate dehydrogenase, a target known to modulate the EBOV replication complex. They also included novel classes of inhibitors, including inhibitors of protein synthesis and hypoxia inducible factor-1. Five compounds were tested for their ability to inhibit replication of a recombinant EBOV that expresses GFP (EBOV-GFP), and four inhibited EBOV-GFP growth at sub-cytotoxic concentrations. These data demonstrate the utility of the HTS minigenome assay for drug discovery and suggest potential directions for antifiloviral drug development.

10.
J Virol ; 89(16): 8346-64, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26041282

ABSTRACT

UNLABELLED: In our study, we characterized the effect of monensin, an ionophore that is known to raise the intracellular pH, on the hepatitis C virus (HCV) life cycle. We showed that monensin inhibits HCV entry in a pangenotypic and dose-dependent manner. Monensin induces an alkalization of intracellular organelles, leading to an inhibition of the fusion step between viral and cellular membranes. Interestingly, we demonstrated that HCV cell-to-cell transmission is dependent on the vesicular pH. Using the selective pressure of monensin, we selected a monensin-resistant virus which has evolved to use a new entry route that is partially pH and clathrin independent. Characterization of this mutant led to the identification of two mutations in envelope proteins, the Y297H mutation in E1 and the I399T mutation in hypervariable region 1 (HVR1) of E2, which confer resistance to monensin and thus allow HCV to use a pH-independent entry route. Interestingly, the I399T mutation introduces an N-glycosylation site within HVR1 and increases the density of virions and their sensitivity to neutralization with anti-apolipoprotein E (anti-ApoE) antibodies, suggesting that this mutation likely induces conformational changes in HVR1 that in turn modulate the association with ApoE. Strikingly, the I399T mutation dramatically reduces HCV cell-to-cell spread. In summary, we identified a mutation in HVR1 that overcomes the vesicular pH dependence, modifies the biophysical properties of particles, and drastically reduces cell-to-cell transmission, indicating that the regulation by HVR1 of particle association with ApoE might control the pH dependence of cell-free and cell-to-cell transmission. Thus, HVR1 and ApoE are critical regulators of HCV propagation. IMPORTANCE: Although several cell surface proteins have been identified as entry factors for hepatitis C virus (HCV), the precise mechanisms regulating its transmission to hepatic cells are still unclear. In our study, we used monensin A, an ionophore that is known to raise the intracellular pH, and demonstrated that cell-free and cell-to-cell transmission pathways are both pH-dependent processes. We generated monensin-resistant viruses that displayed different entry routes and biophysical properties. Thanks to these mutants, we highlighted the importance of hypervariable region 1 (HVR1) of the E2 envelope protein for the association of particles with apolipoprotein E, which in turn might control the pH dependency of cell-free and cell-to-cell transmission.


Subject(s)
Hepacivirus/physiology , Ionophores/pharmacology , Monensin/pharmacology , Viral Envelope Proteins/genetics , Viral Proteins/genetics , Virus Internalization/drug effects , Cell Line , Dose-Response Relationship, Drug , Drug Resistance, Viral/genetics , Fluorescent Antibody Technique, Indirect , Hepacivirus/genetics , Humans , Hydrogen-Ion Concentration/drug effects , Mutation, Missense/genetics , Neutralization Tests , Viral Proteins/metabolism
11.
Nanoscale ; 7(4): 1392-402, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25502878

ABSTRACT

The search for viral entry inhibitors that selectively target viral envelope glycoproteins has attracted increasing interest in recent years. Amongst the handful of molecules reported to show activity as hepatitis C virus (HCV) entry inhibitors are a variety of glycan-binding proteins including the lectins, cyanovirin-N (CV-N) and griffithsin. We recently demonstrated that boronic acid-modified nanoparticles are able to reduce HCV entry through a similar mechanism to that of lectins. A major obstacle to any further development of these nanostructures as viral entry inhibitors is their only moderate maximal inhibition potential. In the present study, we report that lipid nanocapsules (LNCs), surface-functionalized with amphiphilic boronic acid (BA) through their post-insertion into the semi-rigid shell of the LNCs, are indeed far superior as HCV entry inhibitors when compared with previously reported nanostructures. These 2(nd) generation particles (BA-LNCs) are shown to prevent HCV infection in the micromolar range (IC50 = 5.4 µM of BA moieties), whereas the corresponding BA monomers show no significant effects even at the highest analyzed concentration (20 µM). The new BA-LNCs are the most promising boronolectin-based HCV entry inhibitors reported to date and are thus observed to show great promise in the development of a pseudolectin-based therapeutic agent.


Subject(s)
Boronic Acids/chemistry , Hepacivirus/physiology , Nanocapsules/chemistry , Antibodies/immunology , Carbocyanines/chemistry , Cell Line , Cell Survival/drug effects , Humans , Microscopy, Fluorescence , Nanocapsules/toxicity , Particle Size , Polyethylene Glycols/chemistry , Triglycerides/chemistry , Viral Envelope Proteins/immunology , Viral Envelope Proteins/metabolism , Virus Internalization/drug effects
12.
ACS Appl Mater Interfaces ; 5(23): 12488-98, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24180242

ABSTRACT

Phenylboronic-acid-modified nanoparticles (NPs) are attracting considerable attention for biological and biomedical applications. We describe here a convenient and general protocol for attaching multiple copies of para-substituted phenylboronic acid moieties onto either iron-oxide-, silica- or diamond-derived NPs. The boronic acid functionalized NPs are all fabricated by first modifying the surface of each particle type with 4-azidobenzoic ester functions. These azide-terminated nanostructures were then reacted with 4-[1-oxo-4-pentyn-1-yl) amino]phenylboronic acid units via a Cu(I) catalyzed Huisgen cycloaddition to furnish, conveniently, the corresponding boronic-acid modified NPs (or "borono-lectins") targeted in this work. The potential of these novel "borono-lectins" as antiviral inhibitors was investigated against the Hepatitis C virus (HCV) exploiting a bioassay that measures the potential of drugs to interfere with the ability of cell-culture-derived JFH1 virus particles to infect healthy hepatocytes. As far as we are aware, this is the first report that describes NP-derived viral entry inhibitors and thus serves as a "proof-of-concept" study. The novel viral entry activity demonstrated, and the fact that the described boronic-acid-functionalized NPs all display much reduced cellular toxicities compared with alternate NPs, sets the stage for their further investigation. The data supports that NP-derived borono-lectins should be pursued as a potential therapeutic strategy for blocking viral entry of HCV.


Subject(s)
Antiviral Agents/pharmacology , Boronic Acids/chemistry , Nanoparticles , Magnetic Resonance Spectroscopy , Spectroscopy, Fourier Transform Infrared
13.
Hepatology ; 58(1): 86-97, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23348596

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) is a major cause of chronic liver disease. Despite recent success in improving anti-HCV therapy, additional progress is still needed to develop cheaper and interferon (IFN)-free treatments. Here, we report that ferroquine (FQ), an antimalarial ferrocenic analog of chloroquine, is a novel inhibitor of HCV. FQ potently inhibited HCV infection of hepatoma cell lines by affecting an early step of the viral life cycle. The antiviral activity of FQ on HCV entry was confirmed with pseudoparticles expressing HCV envelope glycoproteins E1 and E2 from six different genotypes. In addition to its effect on HCV entry, FQ also inhibited HCV RNA replication, albeit at a higher concentration. We also showed that FQ has no effect on viral assembly and virion secretion. Using a binding assay at 4°C, we showed that FQ does not prevent attachment of the virus to the cell surface. Furthermore, virus internalization was not affected by FQ, whereas the fusion process was impaired in the presence of FQ as shown in a cell-cell fusion assay. Finally, virus with resistance to FQ was selected by sequential passage in the presence of the drug, and resistance was shown to be conferred by a single mutation in E1 glycoprotein (S327A). By inhibiting cell-free virus transmission using a neutralizing antibody, we also showed that FQ inhibits HCV cell-to-cell spread between neighboring cells. Combinations of FQ with IFN, or an inhibitor of HCV NS3/4A protease, also resulted in additive to synergistic activity. CONCLUSION: FQ is a novel, interesting anti-HCV molecule that could be used in combination with other direct-acting antivirals.


Subject(s)
Aminoquinolines/pharmacology , Antiviral Agents/therapeutic use , Ferrous Compounds/pharmacology , Hepacivirus/drug effects , Virus Internalization/drug effects , Virus Replication/drug effects , Cell Line, Tumor , Drug Synergism , Hepacivirus/genetics , Hepatitis C/prevention & control , Humans , Interferon-alpha/administration & dosage , Metallocenes , Proline/administration & dosage , Proline/analogs & derivatives , Viral Envelope Proteins/drug effects , Viral Envelope Proteins/genetics , Viral Nonstructural Proteins/antagonists & inhibitors
14.
Expert Opin Biol Ther ; 10(11): 1563-72, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20932226

ABSTRACT

IMPORTANCE OF THE FIELD: Hepatic cirrhosis is a frequent consequence of chronic hepatitis infection (HBV and HCV) or alcohol abuse and the most common cause of hepatocellular carcinoma (HCC). Currently, liver transplantation remains the only effective therapeutic approach for cirrhosis-related HCC patients. The evolution of the pathology strongly depends on immunological mechanisms. AREAS COVERED IN THIS REVIEW: Despite the presence of specific T cells, viral chronic infection and continuous tumor growth suggest a failure of immune control. It appears that direct suppression of antiviral or antitumor effector cells by regulatory T cells plays a pivotal role in the impairment of immune response. Several types of regulatory T cells have been described, natural regulatory T cells (nTreg) and induced-type 1 regulatory T cells (Tr1) being the best characterized. WHAT THE READER WILL GAIN: Currently, there is no evidence for a direct implication of regulatory T cells in the evolution of hepatitis, especially concerning chronic infection, cirrhosis late stage and HCC progress. However, recent studies show that regulatory T cells are implicated in the modulation of HBV- and HCV-associated immune response, thus, promoting HCC progress. TAKE HOME MESSAGE: Therefore, nTreg and Tr1 cells seem to play an important role in the control of immune response leading to chronic hepatitis infection and progression of the pathology to cirrhosis and HCC.


Subject(s)
Hepatitis B, Chronic/immunology , Hepatitis C, Chronic/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/surgery , Cytokines/metabolism , Disease Progression , Humans , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Interleukin-2/physiology , Interleukin-2 Receptor alpha Subunit/analysis , Liver/immunology , Liver Cirrhosis/etiology , Liver Cirrhosis/immunology , Liver Cirrhosis/surgery , Liver Neoplasms/etiology , Liver Neoplasms/immunology , Liver Neoplasms/surgery , Liver Transplantation , Mice , Mice, Transgenic , Postoperative Complications/immunology , Postoperative Complications/virology , Recurrence , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...