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










Publication year range
1.
Cell Microbiol ; 23(4): e13302, 2021 04.
Article in English | MEDLINE | ID: mdl-33432690

ABSTRACT

With no available therapies, infections with Zika virus (ZIKV) constitute a major public health concern as they can lead to congenital microcephaly. In order to generate an intracellular environment favourable to viral replication, ZIKV induces endomembrane remodelling and the morphogenesis of replication factories via enigmatic mechanisms. In this study, we identified the AAA+ type ATPase valosin-containing protein (VCP) as a cellular interaction partner of ZIKV non-structural protein 4B (NS4B). Importantly, its pharmacological inhibition as well as the expression of a VCP dominant-negative mutant impaired ZIKV replication. In infected cells, VCP is relocalised to large ultrastructures containing both NS4B and NS3, which are reminiscent of dengue virus convoluted membranes. Moreover, short treatment with the VCP inhibitors NMS-873 or CB-5083 drastically decreased the abundance and size of ZIKV-induced convoluted membranes. Furthermore, NMS-873 treatment inhibited ZIKV-induced mitochondria elongation previously reported to be physically and functionally linked to convoluted membranes in case of the closely related dengue virus. Finally, VCP inhibition resulted in enhanced apoptosis of ZIKV-infected cells strongly suggesting that convoluted membranes limit virus-induced cytopathic effects. Altogether, this study identifies VCP as a host factor required for ZIKV life cycle and more precisely, for the maintenance of viral replication factories. Our data further support a model in which convoluted membranes regulate ZIKV life cycle by impacting on mitochondrial functions and ZIKV-induced death signals in order to create a cytoplasmic environment favourable to viral replication.


Subject(s)
Adenosine Triphosphatases/metabolism , Apoptosis , Gene Expression Regulation , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism , Zika Virus/genetics , Zika Virus/physiology , Acetanilides/pharmacology , Adenosine Triphosphatases/genetics , Animals , Benzothiazoles/pharmacology , Cell Line, Tumor , Chlorocebus aethiops , HEK293 Cells , Host Microbial Interactions/drug effects , Host Microbial Interactions/genetics , Humans , Indoles/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/virology , Pyrimidines/pharmacology , Valosin Containing Protein/antagonists & inhibitors , Vero Cells
2.
Sci Rep ; 8(1): 8446, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29855523

ABSTRACT

Activation of the inflammasome pathway is crucial for effective intracellular host defense. The mitochondrial network plays an important role in inflammasome regulation but the mechanisms linking mitochondrial homeostasis to attenuation of inflammasome activation are not fully understood. Here, we report that the Parkinson's disease-associated mitochondrial serine protease HtrA2 restricts the activation of ASC-dependent NLRP3 and AIM2 inflammasomes, in a protease activity-dependent manner. Consistently, disruption of the protease activity of HtrA2 results in exacerbated NLRP3 and AIM2 inflammasome responses in macrophages ex vivo and systemically in vivo. Mechanistically, we show that the HtrA2 protease activity regulates autophagy and controls the magnitude and duration of inflammasome signaling by preventing prolonged accumulation of the inflammasome adaptor ASC. Our findings identify HtrA2 as a non-redundant mitochondrial quality control effector that keeps NLRP3 and AIM2 inflammasomes in check.


Subject(s)
DNA-Binding Proteins/metabolism , High-Temperature Requirement A Serine Peptidase 2/metabolism , Inflammasomes/metabolism , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Autophagy , Bone Marrow Cells/cytology , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/metabolism , DNA-Binding Proteins/antagonists & inhibitors , High-Temperature Requirement A Serine Peptidase 2/deficiency , High-Temperature Requirement A Serine Peptidase 2/genetics , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-1beta/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
3.
Immunity ; 43(4): 751-63, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26384545

ABSTRACT

The crosstalk between inflammation and tumorigenesis is now clearly established. However, how inflammation is elicited in the metastatic environment and the corresponding contribution of innate immunity pathways in suppressing tumor growth at secondary sites are poorly understood. Here, we show that mice deficient in Nlrp3 inflammasome components had exacerbated liver colorectal cancer metastatic growth, which was mediated by impaired interleukin-18 (IL-18) signaling. Control of tumor growth was independent of differential cancer cell colonization or proliferation, intestinal microbiota effects, or tumoricidal activity by the adaptive immune system. Instead, the inflammasome-IL-18 pathway impacted maturation of hepatic NK cells, surface expression of the death ligand FasL, and capacity to kill FasL-sensitive tumors. Our results define a regulatory signaling circuit within the innate immune system linking inflammasome activation to effective NK-cell-mediated tumor attack required to suppress colorectal cancer growth in the liver.


Subject(s)
Adenocarcinoma/secondary , Carrier Proteins/physiology , Colorectal Neoplasms/pathology , Inflammasomes/physiology , Killer Cells, Natural/immunology , Liver Neoplasms/secondary , Adenocarcinoma/immunology , Animals , Apoptosis Regulatory Proteins/deficiency , Calcium-Binding Proteins/deficiency , Caspase 1/deficiency , Cell Line, Tumor , Colorectal Neoplasms/immunology , Cytotoxicity, Immunologic , DNA-Binding Proteins/deficiency , Fas Ligand Protein/physiology , Gastrointestinal Microbiome , Immunity, Innate , Immunologic Surveillance , Inflammasomes/deficiency , Interleukin-18/physiology , Interleukin-1beta/physiology , Liver Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasm Proteins/deficiency , Neoplasm Proteins/physiology , Radiation Chimera , Radiation Tolerance , Tumor Microenvironment
4.
PLoS One ; 9(7): e102605, 2014.
Article in English | MEDLINE | ID: mdl-25033043

ABSTRACT

Although studies have addressed the exhaustion of the host's immune response to HCV and its role in treatment, there is little information about the possible contribution of innate immunity to treatment-induced clearance. We hypothesized that because intact myeloid dendritic cell (MDC) pathogen sensing functions are associated with improved HCV-specific CD8+ T cell functionality in some chronically infected patients, it might enhance HCV clearance rate under standard interferon therapy. To investigate this hypothesis, TLR-induced MDC activation and HCV-specific CD8+ T cell response quality were monitored longitudinally at the single-cell level using polychromatic flow cytometry in chronically infected patients undergoing interferon therapy. We correlated the immunological, biochemical and virological data with response to treatment. We demonstrate that the clinical efficacy of interferon-induced viral clearance is influenced by the extent to which HCV inhibits MDC functions before treatment, rather than solely on a breakdown of the extrinsic T cell immunosuppressive environment. Thus, viral inhibition of MDC functions before treatment emerges as a co-determining factor in the clinical efficacy of interferon therapy during chronic HCV infection.


Subject(s)
Antiviral Agents/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Hepacivirus/immunology , Receptors, Pattern Recognition/immunology , Adult , Drug Therapy, Combination , Female , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/virology , Humans , Immunity, Innate/immunology , Interferon alpha-2 , Interferon-alpha/therapeutic use , Lymphocyte Activation/immunology , Male , Middle Aged , Myeloid Cells/immunology , Polyethylene Glycols/therapeutic use , Recombinant Proteins/therapeutic use , Ribavirin/therapeutic use , Toll-Like Receptors/immunology , Virus Replication/drug effects , Virus Replication/immunology
5.
Cell Host Microbe ; 15(1): 23-35, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24439895

ABSTRACT

Cellular inhibitors of apoptosis proteins (cIAPs) are essential regulators of cell death and immunity. The corresponding contributions of IAPs to infectious disease outcomes are relatively unexplored. We find that mice deficient in cIAP2 exhibit increased susceptibility and mortality to influenza A virus infection. The lethality was not due to impaired antiviral immune functions, but rather because of death-receptor-induced programmed necrosis of airway epithelial cells that led to severe bronchiole epithelial degeneration, despite control of viral replication. Pharmacological inhibition of RIPK1 or genetic deletion of Ripk3, both kinases involved in programmed necrosis, rescued cIAP2-deficient mice from influenza-induced lethality. Genetic deletion of the death receptor agonists Fas ligand or TRAIL from the hematopoietic compartment also reversed the susceptibility of cIAP2-deficient mice. Thus, cIAP2-dependent antagonism of RIPK3-mediated programmed necrosis critically protects the host from influenza infection through maintenance of pulmonary tissue homeostasis rather than through pathogen control by the immune system.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Inhibitor of Apoptosis Proteins/immunology , Lung/immunology , Necrosis/immunology , Orthomyxoviridae Infections/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Respiratory Mucosa/immunology , Animals , Baculoviral IAP Repeat-Containing 3 Protein , Fas Ligand Protein/deficiency , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Gene Expression Regulation , Homeostasis/immunology , Host-Pathogen Interactions , Humans , Immunity, Innate , Inhibitor of Apoptosis Proteins/deficiency , Inhibitor of Apoptosis Proteins/genetics , Lung/pathology , Lung/virology , Mice , Mice, Knockout , Necrosis/complications , Necrosis/genetics , Necrosis/mortality , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/mortality , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Signal Transduction , Survival Analysis , TNF-Related Apoptosis-Inducing Ligand/deficiency , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/immunology , Ubiquitin-Protein Ligases
6.
PLoS One ; 7(2): e31012, 2012.
Article in English | MEDLINE | ID: mdl-22363534

ABSTRACT

Cerebral malaria (CM) is a lethal neurological complication of malaria. We implemented a genome-wide screen in mutagenized mice to identify host proteins involved in CM pathogenesis and whose inhibition may be of therapeutic value. One pedigree (P48) segregated a resistance trait whose CM-protective effect was fully penetrant, mapped to chromosome 8, and identified by genome sequencing as homozygosity for a mis-sense mutation (W81R) in the FERM domain of Janus-associated kinase 3 (Jak3). The causative effect of Jak3(W81R) was verified by complementation testing in Jak3(W81R/-) double heterozygotes that were fully protected against CM. Jak3(W81R) homozygotes showed defects in thymic development with depletion of CD8(+) T cell, B cell, and NK cell compartments, and defective T cell-dependent production of IFN-γ. Adoptive transfer of normal splenocytes abrogates CM resistance in Jak3(W81R) homozygotes, an effect attributed to the CD8(+) T cells. Jak3(W81R) behaves as a dominant negative variant, with significant CM resistance of Jak3(W81R/+) heterozygotes, compared to CM-susceptible Jak3(+/+) and Jak3(+/-) controls. CM resistance in Jak3(W81R/+) heterozygotes occurs in presence of normal T, B and NK cell numbers. These findings highlight the pathological role of CD8(+) T cells and Jak3-dependent IFN-γ-mediated Th1 responses in CM pathogenesis.


Subject(s)
Genes, Dominant/genetics , Janus Kinase 3/genetics , Malaria, Cerebral/enzymology , Malaria, Cerebral/prevention & control , Mutation/genetics , Adoptive Transfer , Amino Acid Sequence , Animals , Chromosomes, Mammalian/genetics , Citrobacter/physiology , Ethylnitrosourea , Female , Genetic Predisposition to Disease , Heterozygote , Homozygote , Immunophenotyping , Janus Kinase 3/chemistry , Malaria, Cerebral/genetics , Malaria, Cerebral/immunology , Male , Mice , Mice, Neurologic Mutants , Molecular Sequence Data , Mycobacterium/physiology , Pedigree , Phenotype , Plasmodium berghei/physiology , Protein Structure, Tertiary , Spleen/pathology
7.
J Hepatol ; 56(1): 70-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21835140

ABSTRACT

BACKGROUND & AIMS: Innate sensing of viral infection activates a global defense response including type I interferon (IFN) and IFN-stimulated genes (ISGs) expression. We previously reported that HCV NS3/4A protease, an essential protein in viral polyprotein processing, can abrogate antiviral signaling pathways and effectors' response when ectopically expressed in human hepatocytes by cleaving antiviral adaptor CARDIF. However, whether HCV mediates evasion of innate immunity in patients with chronic infection remains unclear. METHODS: In this study, paired liver biopsies and corresponding purified hepatocytes of chronic hepatitis C patients and controls were subjected to transcriptional analysis of selected innate immune genes and to CARDIF protein detection. RESULTS: We report that an antiviral response is largely supported by infected hepatocytes as demonstrated by upregulation of the representative antiviral genes ISG15, ISG56, and OASL as well as chemokines genes CXCL9, CXCL10, and CXCL11 measured in both HCV-derived liver biopsies and hepatocytes; that the mRNA levels of these indicator ISGs correlate inversely with HCV RNA level; and more importantly that expression of the early responsive IRF3-dependent genes type I IFNß, type III IL28A/IL29, and chemokine CCL5 are severely compromised and associated to a global decrease of CARDIF adaptor in infected hepatocytes. CONCLUSIONS: Altogether the data argue for a strong viral strategy that counteracts the host's early antiviral response of hepatocytes from chronic patients without impairing ISGs induced via classical IFN pathway.


Subject(s)
Hepatitis C, Chronic/immunology , Immunity, Innate , Liver/immunology , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Case-Control Studies , Chemokines/genetics , Female , Hepatitis C, Chronic/genetics , Hepatitis C, Chronic/metabolism , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Immunity, Innate/genetics , Interferon Regulatory Factors/genetics , Interferons/genetics , Liver/metabolism , Liver/virology , Male , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/metabolism , Up-Regulation
8.
Med Sci (Paris) ; 26(10): 869-74, 2010 Oct.
Article in French | MEDLINE | ID: mdl-20929679

ABSTRACT

Chronic hepatitis C virus (HCV) is a liver-borne infectious disease that remains a major global health threat. The mechanisms whereby HCV evades the host's immune defences and establishes persistent infection remain elusive; but they likely require a complex and coordinated interruption of the interplay between innate and adaptive immune actors. This review discusses the concept that HCV evades the host's immune response to its components partly because of its ability to inactivate the major orchestrator of the adaptive immune response - the DCs. It argues that DCs constitute an immunologically relevant cellular viral host actively targeted by HCV. This targeting disrupts TRIF- and IPS-1-dependent but not MyD88-coupled pathogen recognition receptors (PRR) sensing pathways in these infected cells to foil the networks by which innate immunity to HCV is translated into virus-specific adaptive immune-mediated host resistance. Thus, as a culprit, this cell-specific and numerically restrained DC defect offers a promising field of investigation in which to study and understand the HCV-restricted nature of the deficit in cellular immunity in persistently infected -individuals who have otherwise normal immune functions to unrelated pathogens. In this model, protective immunity is contingent on proper processing and delivery of danger signals by DCs presenting HCV antigens.


Subject(s)
Dendritic Cells/immunology , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Adaptive Immunity , Antigens, Viral/immunology , Dendritic Cells/virology , Disease Reservoirs , Hepatitis C, Chronic/epidemiology , Humans , Immunity, Innate
9.
Eur J Immunol ; 40(3): 643-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20201021

ABSTRACT

Human genetics research has had a great impact on the genesis of the inflammasome field and the treatment of certain inflammasomopathies. The identification of mutations causing rare autoinflammatory syndromes, reproductive wastage disorders and of single nucleotide polymorphisms influencing susceptibility to complex diseases such as vitiligo, sepsis, and Crohn's disease has not only led to the characterization of novel proteins involved in NOD-like receptor-coupled inflammatory signaling pathways but also to greater insights into pathogenic mechanisms.


Subject(s)
Genetic Predisposition to Disease , Immune System Diseases/genetics , Inflammation/genetics , Multiprotein Complexes/genetics , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Humans , Immune System Diseases/immunology , Inflammation/immunology , Multiprotein Complexes/immunology
10.
J Immunol ; 184(6): 3134-44, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20173023

ABSTRACT

Although chronic viral infections have evolved mechanisms to interfere with aspects of pathogen recognition by dendritic cells (DCs), the role that these APCs play in virus-specific T cell exhaustion is unclear. Herein we report that NS3-dependent suppression of Toll/IL-1 domain-containing adapter-inducing IFN-beta- and IFN-beta promoter stimulator-1- but not MyD88-coupled pathogen-recognition receptor-induced synthesis of proinflammatory cytokines (IL-12 and TNF-alpha) from DCs by hepatitis C virus (HCV) is a distinctive feature of a subgroup of chronically infected patients. The result is decreased CD8(+) T cell polyfunctional capacities (production of IFN-gamma, IL-2, TNF-alpha, and CD107a mobilization) that is confined to HCV specificities and that relates to the extent to which HCV inhibits DC responses in infected subjects, despite comparable plasma viral load, helper T cell environments, and inhibitory programmed death 1 receptor/ligand signals. Thus, subjects in whom pathogen-recognition receptor signaling in DCs was intact exhibited enhanced polyfunctionality (i.e., IL-2-secretion and CD107a). In addition, differences between HCV-infected patients in the ability of CD8(+) T cells to activate multiple functions in response to HCV did not apply to CD8(+) T cells specific for other immune-controlled viruses (CMV, EBV, and influenza). Our findings identify reversible virus evasion of DC-mediated innate immunity as an additional important factor that impacts the severity of polyfunctional CD8(+) T cell exhaustion during a chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Dendritic Cells/immunology , Dendritic Cells/virology , Hepacivirus/immunology , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/virology , Immunosuppression Therapy , Antigens, Surface/physiology , Apoptosis Regulatory Proteins/physiology , CD8-Positive T-Lymphocytes/pathology , Cell Degranulation/immunology , Dendritic Cells/pathology , Female , Hepatitis C, Chronic/pathology , Humans , Interleukin-2/biosynthesis , Longitudinal Studies , Male , Middle Aged , Myeloid Cells/immunology , Myeloid Cells/pathology , Myeloid Cells/virology , Programmed Cell Death 1 Receptor , Receptors, Pattern Recognition/antagonists & inhibitors , Receptors, Pattern Recognition/physiology , Viral Load/immunology , Virus Activation/immunology
11.
J Hepatol ; 52(2): 167-75, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20006398

ABSTRACT

BACKGROUND & AIMS: Molecular sensors recognize viral nucleic acids and initiate events that subsequently enable cells to control and clear infection. Hepatitis C Virus (HCV) can interfere with the innate host response and the NS3/4A protease was reported to specifically block antiviral signaling pathways, a finding that had yet to be studied in human primary hepatocytes. METHODS: Freshly isolated human primary hepatocytes, transduced with a lentiviral vector expressing HCV NS3/4A were stimulated with extracellular and intracellular double-stranded RNA (dsRNA) and the innate immune antiviral genes were quantified by quantitative PCR and microarrays analysis. RESULTS: We demonstrate that sensing receptors of human hepatocytes in primary cultures are stimulated following recognition of either mode of dsRNA delivery, inducing transcriptional up-regulation (over 100-fold) of multiple immune genes, either selectively or independently of recognition pathways. We also report that the intracellular dsRNA-activated innate response is severely compromised upon ectopic expression of the HCV NS3/4A protease gene in normal human primary hepatocytes, and completely restored by treatment with the NS3/4A protease specific inhibitor BILN2061. CONCLUSIONS: The present study indicates that NS3/4A has a wider protease-dependent effect on the intracellular Pathogen Recognition Receptor (PRR)-mediated immune response than on its extracellular counterpart, which underlies the major role of cytosolic dsRNA receptors in HCV recognition by primary human hepatocytes.


Subject(s)
Hepatocytes/metabolism , Hepatocytes/virology , Viral Nonstructural Proteins/metabolism , Cells, Cultured , Gene Expression Profiling , Hepacivirus/immunology , Hepacivirus/metabolism , Hepatocytes/immunology , Humans , Immunity, Innate , In Vitro Techniques , Oligonucleotide Array Sequence Analysis , RNA/genetics , RNA/metabolism , Signal Transduction
12.
BMC Cancer ; 8: 337, 2008 Nov 17.
Article in English | MEDLINE | ID: mdl-19014680

ABSTRACT

BACKGROUND: The Ras-dependent ERK1/2 MAP kinase signaling pathway plays a central role in cell proliferation control and is frequently activated in human colorectal cancer. Small-molecule inhibitors of MEK1/MEK2 are therefore viewed as attractive drug candidates for the targeted therapy of this malignancy. However, the exact contribution of MEK1 and MEK2 to the pathogenesis of colorectal cancer remains to be established. METHODS: Wild type and constitutively active forms of MEK1 and MEK2 were ectopically expressed by retroviral gene transfer in the normal intestinal epithelial cell line IEC-6. We studied the impact of MEK1 and MEK2 activation on cellular morphology, cell proliferation, survival, migration, invasiveness, and tumorigenesis in mice. RNA interference was used to test the requirement for MEK1 and MEK2 function in maintaining the proliferation of human colorectal cancer cells. RESULTS: We found that expression of activated MEK1 or MEK2 is sufficient to morphologically transform intestinal epithelial cells, dysregulate cell proliferation and induce the formation of high-grade adenocarcinomas after orthotopic transplantation in mice. A large proportion of these intestinal tumors metastasize to the liver and lung. Mechanistically, activation of MEK1 or MEK2 up-regulates the expression of matrix metalloproteinases, promotes invasiveness and protects cells from undergoing anoikis. Importantly, we show that silencing of MEK2 expression completely suppresses the proliferation of human colon carcinoma cell lines, whereas inactivation of MEK1 has a much weaker effect. CONCLUSION: MEK1 and MEK2 isoforms have similar transforming properties and are able to induce the formation of metastatic intestinal tumors in mice. Our results suggest that MEK2 plays a more important role than MEK1 in sustaining the proliferation of human colorectal cancer cells.


Subject(s)
Adenocarcinoma/secondary , Cell Transformation, Neoplastic , Intestinal Mucosa/pathology , Intestinal Neoplasms/pathology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Animals , Anoikis , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Humans , Intestinal Mucosa/metabolism , Intestinal Neoplasms/enzymology , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 2/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Protein Isoforms/metabolism , RNA Interference , Rats
13.
J Virol ; 81(11): 5537-46, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17376921

ABSTRACT

The role of peripheral dendritic cells (DCs) in hepatitis C virus (HCV) infection is unclear. To determine if persistent infection exerts an inhibitory pressure on HCV-specific innate responses, we analyzed DC function in blood through quantification of cell-associated HCV RNA levels in conjunction with multiparametric flow cytometry analysis of pathogen recognition receptor-induced cytokine expression. Independently of the serum viral load, fluorescence-activated cell sorter-purified total DCs had a wide range of cell-associated HCV genomic RNA copy numbers (mean log(10), 5.0 per 10(6) cells; range, 4.3 to 5.8). Here we report that for viremic patients with high viral loads in their total DCs, the myeloid DC (MDC) subset displayed impaired expression of interleukin-12 (IL-12) and tumor necrosis factor alpha (TNF-alpha) but normal IL-6 or chemokine CCL3 expression in response to poly(I:C) and lipopolysaccharide (LPS). IL-6-expressing cells from this subgroup of viremic patients demonstrated a significant increase (sixfold more) in TNF-alpha(-) IL-12(-) cell frequency compared to healthy donors (mean, 38.8% versus 6.5%; P < 0.0001), indicating a functional defect in a subpopulation of cytokine-producing MDCs ( approximately 6% of MDCs). Attenuation of poly(I:C) and LPS innate sensing was HCV RNA density dependent and did not correlate with viremia or deficits in circulating MDC frequencies in HCV-infected patients. Monocytes from these patients were functionally intact, responding normally on a per-cell basis following stimulation, independent of cell-associated HCV RNA levels. Taken together, these data indicate that detection of HCV genomic RNA in DCs and loss of function in the danger signal responsiveness of a small proportion of DCs in vivo are interrelated rather than independent phenomena.


Subject(s)
Dendritic Cells/metabolism , Hepacivirus/physiology , Hepatitis C, Chronic/blood , Lipopolysaccharides/metabolism , Myeloid Cells/metabolism , Poly I-C/metabolism , Adult , Aged , Cells, Cultured , Dendritic Cells/pathology , Dendritic Cells/virology , Female , Hepacivirus/genetics , Hepatitis C, Chronic/pathology , Hepatitis C, Chronic/virology , Humans , Male , Middle Aged , Myeloid Cells/pathology , RNA, Viral/blood
14.
Proc Natl Acad Sci U S A ; 100(5): 2783-8, 2003 Mar 04.
Article in English | MEDLINE | ID: mdl-12594341

ABSTRACT

RNA interference represents an exciting new technology that could have therapeutic applications for the treatment of viral infections. Hepatitis C virus (HCV) is a major cause of chronic liver disease and affects >270 million individuals worldwide. The HCV genome is a single-stranded RNA that functions as both a messenger RNA and replication template, making it an attractive target for the study of RNA interference. Double-stranded small interfering RNA (siRNA) molecules designed to target the HCV genome were introduced through electroporation into a human hepatoma cell line (Huh-7) that contained an HCV subgenomic replicon. Two siRNAs dramatically reduced virus-specific protein expression and RNA synthesis to levels that were 90% less than those seen in cells treated with negative control siRNAs. These same siRNAs protected naive Huh-7 cells from challenge with HCV replicon RNA. Treatment of cells with synthetic siRNA was effective >72 h, but the duration of RNA interference could be extended beyond 3 weeks through stable expression of complementary strands of the interfering RNA by using a bicistronic expression vector. These results suggest that a gene-therapeutic approach with siRNA could ultimately be used to treat HCV.


Subject(s)
Hepatitis C/metabolism , Liver/cytology , RNA Interference , RNA, Small Interfering/physiology , RNA, Viral/genetics , RNA/metabolism , Virus Replication/physiology , Antibodies, Monoclonal/metabolism , Blotting, Northern , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Electroporation , Genetic Vectors , Humans , Models, Genetic , Mutation , Plasmids/metabolism , RNA, Messenger/metabolism , Time Factors , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Virus Replication/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...