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










Publication year range
1.
Oncotarget ; 9(31): 21978-22000, 2018 Apr 24.
Article in English | MEDLINE | ID: mdl-29774117

ABSTRACT

Tumor-tumor distant interactions within one organism are of major clinical relevance determining clinical outcome. To investigate this poorly understood phenomenon, a double human cervical xenograft model in nude mice was developed. A first tumor was induced subcutaneously by injection of human papillomavirus positive cervical carcinoma cells into the mouse lower right flank and 3 weeks later, animals were challenged with the same tumor cell line injected subcutaneously into the upper left flank. These tumors had no direct physical contact and we found no systemic changes induced by the primary tumor affecting the growth of a secondary tumor. However, ablation of the primary tumor by local treatment with cidofovir, a nucleotide analogue with known antiviral and antiproliferative properties, resulted not only in a local antitumor effect but also in a temporary far-reaching effect leading to retarded growth of the challenged tumor. Cidofovir far-reaching effects were linked to a reduced tumor-driven inflammation, to increased anti-tumor immune responses, and could not be enhanced by co-administration with immune stimulating adjuvants. Our findings point to the potential use of cidofovir in novel therapeutic strategies aiming to kill tumor cells as well as to influence the immune system to fight cancer.

2.
Front Immunol ; 9: 364, 2018.
Article in English | MEDLINE | ID: mdl-29535732

ABSTRACT

Recent studies have reported that the crosslinking of regulatory receptors (RRs), such as blood dendritic cell antigen 2 (BDCA-2) (CD303) or ILT7 (CD85g), of plasmacytoid dendritic cells (pDCs) efficiently suppresses the production of type I interferons (IFN-I, α/ß/ω) and other cytokines in response to toll-like receptor 7 and 9 (TLR7/9) ligands. The exact mechanism of how this B cell receptor (BCR)-like signaling blocks TLR7/9-mediated IFN-I production is unknown. Here, we stimulated BCR-like signaling by ligation of RRs with BDCA-2 and ILT7 mAbs, hepatitis C virus particles, or BST2 expressing cells. We compared BCR-like signaling in proliferating pDC cell line GEN2.2 and in primary pDCs from healthy donors, and addressed the question of whether pharmacological targeting of BCR-like signaling can antagonize RR-induced pDC inhibition. To this end, we tested the TLR9-mediated production of IFN-I and proinflammatory cytokines in pDCs exposed to a panel of inhibitors of signaling molecules involved in BCR-like, MAPK, NF-ĸB, and calcium signaling pathways. We found that MEK1/2 inhibitors, PD0325901 and U0126 potentiated TLR9-mediated production of IFN-I in GEN2.2 cells. More importantly, MEK1/2 inhibitors significantly increased the TLR9-mediated IFN-I production blocked in both GEN2.2 cells and primary pDCs upon stimulation of BCR-like or phorbol 12-myristate 13-acetate-induced protein kinase C (PKC) signaling. Triggering of BCR-like and PKC signaling in pDCs resulted in an upregulation of the expression and phoshorylation of c-FOS, a downstream gene product of the MEK1/2-ERK pathway. We found that the total level of c-FOS was higher in proliferating GEN2.2 cells than in the resting primary pDCs. The PD0325901-facilitated restoration of the TLR9-mediated IFN-I production correlated with the abrogation of MEK1/2-ERK-c-FOS signaling. These results indicate that the MEK1/2-ERK pathway inhibits TLR9-mediated type I IFN production in pDCs and that pharmacological targeting of MEK1/2-ERK signaling could be a strategy to overcome immunotolerance of pDCs and re-establish their immunogenic activity.


Subject(s)
B-Lymphocytes/immunology , Dendritic Cells/physiology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Receptors, Antigen, B-Cell/metabolism , Calcium Signaling , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interferon Type I/metabolism , MAP Kinase Signaling System , NF-kappa B/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Antigen, B-Cell/genetics , Toll-Like Receptor 9/metabolism
3.
J Transl Med ; 16(1): 1, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29316942

ABSTRACT

BACKGROUND: Plasma extracellular vesicles (EVs), especially exosome-like vesicles (ELVs), are being increasingly explored as a source of potential noninvasive disease biomarkers. The discovery of blood-based biomarkers associated with ELVs requires methods that isolate high yields of these EVs without significant contamination with highly abundant plasma proteins and lipoproteins. The rising interest in blood-based EV-associated biomarkers has led to the rapid development of novel EV isolation methods. However, the field suffers from a lack of standardization and often, new techniques are used without critical evaluation. Size exclusion chromatography (SEC) has become the method of choice for rapid isolation of relatively pure EVs from plasma, yet it has technical limitations for certain downstream applications. The recently released exoEasy kit (Qiagen) is a new membrane affinity spin column method for the isolation of highly pure EVs from biofluids with the potential to overcome most of the limitations of SEC. METHODS: By using multiple complementary techniques we assessed the performance of the exoEasy kit in isolating ELVs from 2 ml of human plasma and compared it with the SEC qEV column (Izon Science). RESULTS: Our data show that exoEasy kit isolates a heterogenous mixture of particles with a larger median diameter, broader size range and a higher yield than the SEC qEV column. The exclusive presence of small RNAs in the particles and the total RNA yield were comparable to the SEC qEV column. Despite being less prone to low density lipoprotein contamination than the SEC qEV column, the overall purity of exoEasy kit EV preparations was suboptimal. The low particle-protein ratio, significant amount of albumin, very low levels of exosome-associated proteins and propensity to triglyceride-rich lipoprotein contamination suggest isolation of mainly non-ELVs and co-isolation of plasma proteins and certain lipoproteins by the exoEasy kit. CONCLUSIONS: We demonstrate that performance of exoEasy kit for the isolation of ELVs for biomarker discovery is inferior to the SEC qEV column. This comprehensive evaluation of a novel EV isolation method contributes to the acceleration of the discovery of EV-associated biomarkers and the development of EV-based diagnostics.


Subject(s)
Chromatography, Affinity/methods , Chromatography, Gel/methods , Exosomes/metabolism , Plasma/metabolism , Blood Proteins/metabolism , Exosomes/ultrastructure , Humans , Lipoproteins/metabolism , Membranes , Nanoparticles/chemistry , RNA/metabolism
4.
Front Immunol ; 8: 394, 2017.
Article in English | MEDLINE | ID: mdl-28439271

ABSTRACT

The innate immune cells sense microbial infection and self-ligands by pathogen recognition receptors (PRRs), such as toll-like receptors (TLRs) and regulatory receptors (RRs), associated with immunoreceptor tyrosine-based activation motif (ITAM). Rapid activation and concerted action of PRRs signaling and feedback inhibitory mechanisms must be engaged to ensure the host defense functions and to prevent cytotoxicity associated with excessive activation. ITAM-associated RRs can generate stimulatory or, paradoxically, inhibitory signals. The network of ITAM-associated RR, together with TLR-signaling pathways, are responsible for immunogenic or tolerogenic responses of macrophages and dendritic cells to their microenvironment. In macrophages, TLR4 signaling is inhibited by low-avidity ligation of ITAM-associated receptors, while high-avidity ligation of ITAM-associated receptors results in potentiation of TLR4 signaling together with resistance to extracellular cytokine microenvironment signals. In contrast to macrophages, TLR7/9 signaling in plasmacytoid DCs (pDCs) is inhibited by high-avidity ligation of ITAM-associated RR, while low-avidity ligation does not show any effect. Surprisingly, interference of ITAM-associated receptor signaling with TLR pathways has not been reported in conventional dendritic cells. Here, we present an overview of molecular mechanisms acting at the crossroads of TLR and ITAM-signaling pathways and address the question of how the high-avidity engagement of the ITAM-associated receptors in pDCs inhibits TLR7/9 signaling. Cellular context and spatiotemporal engagement of ITAM- and TLR-signaling pathways are responsible for different outcomes of macrophage versus pDC activation. While the cross-regulation of cytokine and TLR signaling, together with antigen presentation, are the principal functions of ITAM-associated RR in macrophages, the major role of these receptors in pDCs seems to be related to inhibition of cytokine production and reestablishment of a tolerogenic state following pDC activation. Pharmacologic targeting of TLR and ITAM signaling could be an attractive new therapeutic approach for treatment of chronic infections, cancer, and autoimmune and inflammatory diseases related to pDCs.

5.
World J Hepatol ; 8(21): 902-14, 2016 Jul 28.
Article in English | MEDLINE | ID: mdl-27478540

ABSTRACT

AIM: To evaluate the antiviral potency of a new anti-hepatitis C virus (HCV) antiviral agent targeting the cellular autophagy machinery. METHODS: Non-infected liver slices, obtained from human liver resection and cut in 350 µm-thick slices (2.7 × 10(6) cells per slice) were infected with cell culture-grown HCV Con1b/C3 supernatant (multiplicity of infection = 0.1) cultivated for up to ten days. HCV infected slices were treated at day 4 post-infection with GNS-396 for 6 d at different concentrations. HCV replication was evaluated by strand-specific real-time quantitative reverse transcription - polymerase chain reaction. The infectivity titers of supernatants were evaluated by foci formation upon inoculation into naive Huh-7.5.1 cells. The cytotoxic effect of the drugs was evaluated by lactate dehydrogenase leakage assays. RESULTS: The antiviral efficacy of a new antiviral drug, GNS-396, an autophagy inhibitor, on HCV infection of adult human liver slices was evidenced in a dose-dependent manner. At day 6 post-treatment, GNS-396 EC50 was 158 nmol/L without cytotoxic effect (compared to hydroxychloroquine EC50 = 1.17 µmol/L). CONCLUSION: Our results demonstrated that our ex vivo model is efficient for evaluation the potency of autophagy inhibitors, in particular a new quinoline derivative GNS-396 as antiviral could inhibit HCV infection in a dose-dependent manner without cytotoxic effect.

6.
PLoS One ; 11(6): e0156063, 2016.
Article in English | MEDLINE | ID: mdl-27258042

ABSTRACT

Crosslinking of regulatory immunoreceptors (RR), such as BDCA-2 (CD303) or ILT7 (CD85g), of plasmacytoid dendritic cells (pDCs) efficiently suppresses production of type-I interferon (IFN)-α/ß and other cytokines in response to Toll-like receptor (TLR) 7/9 ligands. This cytokine-inhibitory pathway is mediated by spleen tyrosine kinase (Syk) associated with the ITAM-containing adapter of RR. Here we demonstrate by pharmacological targeting of Syk that in addition to the negative regulation of TLR7/9 signaling via RR, Syk also positively regulates the TLR7/9 pathway in human pDCs. Novel highly specific Syk inhibitor AB8779 suppressed IFN-α, TNF-α and IL-6 production induced by TLR7/9 agonists in primary pDCs and in the pDC cell line GEN2.2. Triggering of TLR9 or RR signaling induced a differential kinetics of phosphorylation at Y352 and Y525/526 of Syk and a differential sensitivity to AB8779. Consistent with the different roles of Syk in TLR7/9 and RR signaling, a concentration of AB8779 insufficient to block TLR7/9 signaling still released the block of IFN-α production triggered via the RR pathway, including that induced by hepatitis B and C viruses. Thus, pharmacological targeting of Syk partially restored the main pDC function-IFN-α production. Opposing roles of Syk in TLR7/9 and RR pathways may regulate the innate immune response to weaken inflammation reaction.


Subject(s)
Dendritic Cells/metabolism , Signal Transduction/physiology , Syk Kinase/metabolism , Toll-Like Receptors/metabolism , Cytokines/metabolism , Dendritic Cells/drug effects , Humans , Immunity, Innate , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Syk Kinase/antagonists & inhibitors , Toll-Like Receptors/agonists
7.
Oncotarget ; 7(30): 47302-47318, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-27331622

ABSTRACT

Human papillomavirus (HPV) causes cervical cancer and a large fraction of head and neck squamous cell carcinomas (HNSCC). Cidofovir (CDV) proved efficacious in the treatment of several HPV-induced benign and malignant hyper proliferations. To provide a better insight into how CDV selectively eradicates transformed cells, HPV+ and HPV- cervical carcinoma and HNSCC cell lines were compared to normal cells for antiproliferative effects, CDV metabolism, drug incorporation into cellular DNA, and DNA damage. Incorporation of CDV into cellular DNA was higher in tumor cells than in normal cells and correlated with CDV antiproliferative effects, which were independent of HPV status. Increase in phospho-ATM levels was detected following CDV exposure and higher levels of γ-H2AX (a quantitative marker of double-strand breaks) were measured in tumor cells compared to normal cells. A correlation between DNA damage and CDV incorporation into DNA was found but not between DNA damage and CDV antiproliferative effects. These data indicate that CDV antiproliferative effects result from incorporation of the drug into DNA causing DNA damage. However, the anti-tumor effects of CDV cannot be exclusively ascribed to DNA damage. Furthermore, CDV can be considered a promising broad spectrum anti-cancer agent, not restricted to HPV+ lesions.


Subject(s)
Antiviral Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cytosine/analogs & derivatives , DNA Damage , Head and Neck Neoplasms/drug therapy , Organophosphonates/pharmacology , Papillomaviridae/isolation & purification , Uterine Cervical Neoplasms/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Cell Line, Tumor , Cidofovir , Cytosine/pharmacology , Female , HeLa Cells , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/virology , Humans , Male , Papillomavirus Infections/drug therapy , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Squamous Cell Carcinoma of Head and Neck , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology
8.
Blood ; 120(23): 4544-51, 2012 Nov 29.
Article in English | MEDLINE | ID: mdl-23053572

ABSTRACT

The elimination of hepatitis C virus (HCV) in > 50% of chronically infected patients by treatment with IFN-α suggests that plasmacytoid dendritic cells (pDCs), major producers of IFN-α, play an important role in the control of HCV infection. However, despite large amounts of Toll-like receptor 7-mediated IFN-α, produced by pDCs exposed to HCV-infected hepatocytes, HCV still replicates in infected liver. Here we show that HCV envelope glycoprotein E2 is a novel ligand of pDC C-type lectin immunoreceptors (CLRs), blood DC antigen 2 (BDCA-2) and DC-immunoreceptor (DCIR). HCV particles inhibit, via binding of E2 glycoprotein to CLRs, production of IFN-α and IFN-λ in pDCs exposed to HCV-infected hepatocytes, and induce in pDCs a rapid phosphorylation of Akt and Erk1/2, in a manner similar to the crosslinking of BDCA-2 or DCIR. Blocking of BDCA-2 and DCIR with Fab fragments of monoclonal antibodies preserves the capacity of pDCs to produce type I and III IFNs in the presence of HCV particles. Thus, negative interference of CLR signaling triggered by cell-free HCV particles with Toll-like receptor signaling triggered by cell-associated HCV results in the inhibition of the principal pDC function, production of IFN.


Subject(s)
Dendritic Cells/immunology , Interferons/immunology , Lectins, C-Type/immunology , Membrane Glycoproteins/immunology , Receptors, Immunologic/immunology , Viral Envelope Proteins/immunology , Animals , COS Cells , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Dendritic Cells/metabolism , Dendritic Cells/virology , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Hepacivirus/immunology , Hepacivirus/metabolism , Hepacivirus/physiology , Host-Pathogen Interactions/immunology , Humans , Interferons/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Ligands , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Liver Neoplasms/virology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Viral Envelope Proteins/metabolism
9.
J Virol ; 86(2): 1090-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22090103

ABSTRACT

Plasmacytoid dendritic cells (pDCs) respond to viral infection by production of alpha interferon (IFN-α), proinflammatory cytokines, and cell differentiation. The elimination of hepatitis C virus (HCV) in more than 50% of chronically infected patients by treatment with IFN-α suggests that pDCs can play an important role in the control of HCV infection. pDCs exposed to HCV-infected hepatoma cells, in contrast to cell-free HCV virions, produce large amounts of IFN-α. To further investigate the molecular mechanism of HCV sensing, we studied whether exposure of pDCs to HCV-infected hepatoma cells activates, in parallel to interferon regulatory factor 7 (IRF7)-mediated production of IFN-α, nuclear factor kappa B (NF-κB)-dependent pDC responses, such as expression of the differentiation markers CD40, CCR7, CD86, and tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and secretion of the proinflammatory cytokines TNF-α and interleukin 6 (IL-6). We demonstrate that exposure of pDCs to HCV-infected hepatoma cells surprisingly did not induce phosphorylation of NF-κB or cell surface expression of CD40, CCR7, CD86, or TRAIL or secretion of TNF-α and IL-6. In contrast, CpG-A and CpG-B induced production of TNF-α and IL-6 in pDCs exposed to the HCV-infected hepatoma cells, showing that cell-associated virus did not actively inhibit Toll-like receptor (TLR)-mediated NF-κB phosphorylation. Our results suggest that cell-associated HCV signals in pDCs via an endocytosis-dependent mechanism and IRF7 but not via the NF-κB pathway. In spite of IFN-α induction, cell-associated HCV does not induce a full functional response of pDCs. These findings contribute to the understanding of evasion of immune responses by HCV.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/virology , Hepacivirus/physiology , Hepatitis C/immunology , NF-kappa B/immunology , Signal Transduction , Cell Line, Tumor , Cells, Cultured , Hepacivirus/genetics , Hepatitis C/genetics , Hepatitis C/virology , Humans , Interferon-alpha/genetics , Interferon-alpha/immunology , NF-kappa B/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
10.
Cell Host Microbe ; 6(1): 68-80, 2009 Jul 23.
Article in English | MEDLINE | ID: mdl-19616766

ABSTRACT

Primate lentiviruses, including HIV-1, transduce terminally differentiated, nondividing myeloid cells; however, these cells are refractory to infection by gammaretroviruses such as murine leukemia virus (MLV). Here, we present evidence that a cellular restriction is the obstacle to transduction of macrophages by MLV. Neutralization of the restriction by Vpx, a primate lentiviral protein previously shown to protect primate lentiviruses from a macrophage restriction, rendered macrophages permissive to MLV infection. We further demonstrate that this restriction prevents transduction of quiescent monocytes by HIV-1. Monocyte-HeLa heterokaryons were resistant to HIV-1 infection, while heterokaryons formed between monocytes and HeLa cells expressing Vpx were permissive to HIV-1 infection. Encapsidation of Vpx within HIV-1 virions conferred the ability to infect quiescent monocytes. Collectively, our results indicate that the relative ability of lentiviruses and gammaretroviruses to transduce nondividing myeloid cells is dependent upon their ability to neutralize a cellular restriction.


Subject(s)
HIV-1/growth & development , HIV-1/immunology , Leukemia Virus, Murine/growth & development , Leukemia Virus, Murine/immunology , Macrophages/immunology , Monocytes/immunology , Cell Line , Cells, Cultured , Humans , Macrophages/virology , Monocytes/virology , Transduction, Genetic , Viral Regulatory and Accessory Proteins/immunology
11.
Nat Biotechnol ; 26(10): 1187-92, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18806783

ABSTRACT

The HIV-1 protein Vif, essential for in vivo viral replication, targets the human DNA-editing enzyme, APOBEC3G (A3G), which inhibits replication of retroviruses and hepatitis B virus. As Vif has no known cellular homologs, it is an attractive, yet unrealized, target for antiviral intervention. Although zinc chelation inhibits Vif and enhances viral sensitivity to A3G, this effect is unrelated to the interaction of Vif with A3G. We identify a small molecule, RN-18, that antagonizes Vif function and inhibits HIV-1 replication only in the presence of A3G. RN-18 increases cellular A3G levels in a Vif-dependent manner and increases A3G incorporation into virions without inhibiting general proteasome-mediated protein degradation. RN-18 enhances Vif degradation only in the presence of A3G, reduces viral infectivity by increasing A3G incorporation into virions and enhances cytidine deamination of the viral genome. These results demonstrate that the HIV-1 Vif-A3G axis is a valid target for developing small molecule-based new therapies for HIV infection or for enhancing innate immunity against viruses.


Subject(s)
Anti-HIV Agents/therapeutic use , Cytidine Deaminase/immunology , Cytosine Deaminase/antagonists & inhibitors , Cytosine Deaminase/immunology , HIV Infections/immunology , HIV Infections/prevention & control , Immunity, Innate/immunology , vif Gene Products, Human Immunodeficiency Virus/immunology , APOBEC-3G Deaminase , Anti-HIV Agents/immunology , Cytidine Deaminase/genetics , HIV-1/drug effects , HIV-1/physiology , Humans , Immunity, Innate/drug effects
12.
PLoS Pathog ; 4(5): e1000057, 2008 May 02.
Article in English | MEDLINE | ID: mdl-18451984

ABSTRACT

Primate lentiviruses encode four "accessory proteins" including Vif, Vpu, Nef, and Vpr/Vpx. Vif and Vpu counteract the antiviral effects of cellular restrictions to early and late steps in the viral replication cycle. We present evidence that the Vpx proteins of HIV-2/SIV(SM) promote virus infection by antagonizing an antiviral restriction in macrophages. Fusion of macrophages in which Vpx was essential for virus infection, with COS cells in which Vpx was dispensable for virus infection, generated heterokaryons that supported infection by wild-type SIV but not Vpx-deleted SIV. The restriction potently antagonized infection of macrophages by HIV-1, and expression of Vpx in macrophages in trans overcame the restriction to HIV-1 and SIV infection. Vpx was ubiquitylated and both ubiquitylation and the proteasome regulated the activity of Vpx. The ability of Vpx to counteract the restriction to HIV-1 and SIV infection was dependent upon the HIV-1 Vpr interacting protein, damaged DNA binding protein 1 (DDB1), and DDB1 partially substituted for Vpx when fused to Vpr. Our results indicate that macrophage harbor a potent antiviral restriction and that primate lentiviruses have evolved Vpx to counteract this restriction.


Subject(s)
DNA-Binding Proteins/metabolism , HIV-2/physiology , Macrophages/virology , Viral Regulatory and Accessory Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Gene Expression Regulation, Viral , Gene Silencing , HIV-2/pathogenicity , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics
13.
Int J Antimicrob Agents ; 25(5): 419-26, 2005 May.
Article in English | MEDLINE | ID: mdl-15848298

ABSTRACT

Since some antiviral drugs have a broad spectrum of action, the aim of this study was to assess whether o-(acetoxyphenyl)hept-2-ynyl sulphide (APHS), a recently described inhibitor of human immunodeficiency virus type 1 (HIV-1) replication, has an effect on the replication of other retroviruses, (-) and (+) RNA viruses and DNA viruses. APHS did not affect the replication of feline immunodeficiency virus, HIV-2 and a HIV-1 strain resistant to non-nucleoside reverse transcriptase inhibitors (NNRTI). APHS could also not inhibit the replication of the RNA viruses, respiratory syncytium virus or mouse hepatitis virus. In contrast, APHS did inhibit the replication of wild-type herpes simplex virus type 1 (HSV-1) as well as acyclovir-resistant HSV-1 and HSV-2 mutant. These results suggest that APHS is a NNRTI of HIV-1 replication, but not HIV-2 replication, and that APHS is an inhibitor of both HSV-1 and HSV-2 replication.


Subject(s)
Heptanes/pharmacology , Lentivirus/drug effects , Sulfides/pharmacology , Acyclovir/pharmacology , Alkynes , Animals , Cell Line , Dose-Response Relationship, Drug , Drug Resistance, Bacterial , HIV-1/drug effects , HIV-2/drug effects , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/drug effects , Herpesvirus 2, Human/genetics , Humans , Monocytes/virology , Murine hepatitis virus/drug effects , Mutation , Respiratory Syncytial Viruses/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Virus Replication/drug effects
14.
J Clin Virol ; 32(1): 7-18, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15572000

ABSTRACT

BACKGROUND: Widespread and frequent use of acyclovir (ACV) for treatment, suppressive therapy and prophylaxis of herpes simplex virus (HSV) infections and its over the counter availability may be associated with emergence of HSV resistance. OBJECTIVES: To determine the prevalence of ACV-resistant HSV isolates in different patient groups between 1999 and 2002 in the Netherlands. STUDY DESIGN: A total of 542 isolates, 410 HSV-1 and 132 HSV-2, from 496 patients were screened for reduced susceptibility to ACV. A newly developed ELVIRA HSV screening assay was used that allowed a high throughput screening. The genotypic analysis of the HSV thymidine kinase gene was performed to identify resistance-associated mutations. RESULTS: Thirteen isolates, 8 HSV-1 and 5 HSV-2, from 10 patients (2%) were found resistant to ACV. A single ACV-resistant strain was identified among isolates from 368 immunocompetent patients (0.27%; 95% confidence interval [CI], 0.007%-1.5%), whereas in nine isolates from 128 immunocompromised patients resistant HSV was identified (7%; 95% CI, 3.26%-12.93%). The highest frequency of ACV-resistant HSV was associated with bone marrow transplantation: four patients out of 28 (14.3%) shed resistant virus. In addition, resistant virus was obtained from two HIV-positive patients, one patient with a hematological malignancy and two patients on immunosuppressive drugs. Further testing showed that none of the isolates was resistant to foscarnet. Several new mutations were identified in the thymidine kinase gene of these resistant isolates, and their effect on ACV-resistance is discussed. CONCLUSIONS: Our study shows that the prevalence of ACV resistance is low in immunocompetent patients (0.27%), whereas ACV-resistant HSV infections occur relatively frequently in immunocompromised patients (7%; P < 0.0001). This emphasizes the need for drug susceptibility monitoring of HSV infections in immunocompromised patients with persisting infections despite antiviral therapy.


Subject(s)
Acyclovir/pharmacology , Drug Resistance, Viral , Herpes Simplex/virology , Simplexvirus/drug effects , Acyclovir/therapeutic use , Adult , Female , Herpes Simplex/drug therapy , Humans , Immunocompetence , Male , Microbial Sensitivity Tests , Netherlands/epidemiology , Prevalence , Simplexvirus/classification , Simplexvirus/genetics
15.
Antivir Ther ; 9(4): 565-75, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15456088

ABSTRACT

Thirty-one herpes simplex virus type one (HSV-1) isolates from 12 haematopoietic stem cell transplant recipients with persistent HSV infections despite acyclovir (ACV) prophylaxis or treatment, were genotypically and phenotypically characterized. The relationship between drug susceptibility of the isolates and mutations in thymidine kinase (TK) and DNA polymerase (DNA pol) genes was examined. In all 12 patients, HSV infections were due to ACV-resistant, foscarnet-sensitive viruses. Out of 31 isolates examined, 23 were resistant and eight were sensitive to ACV; eight patients carried viruses with frameshift mutations in the TK gene (due to addition or deletion of single nucleotides in homopolymeric repeats). These mutations were found at codon 61 (G deletion, one patient), 146 (G insertion, five patients) and 153 or 185 (C deletion, one patient each). In four patients, viruses were selected during ACV therapy that contained novel amino acid substitutions in the TK gene (H58R, G129D, A189V, R216H, R220C). Their possible role in ACV resistance was further confirmed phenotypically and by the absence of any resistance-associated mutations in the DNA pol gene. These substitutions were located in ATP- or nucleoside-binding sites or in conserved regions of the TK gene. In addition, a single mutation, Q570R, in the delta-region C of the DNA pol gene, was identified in an isolate from a single patient with resistance to ACV. Our study confirms and expands previous data on genotypic changes associated with ACV resistance of HSV-1 clinical isolates.


Subject(s)
Acyclovir/therapeutic use , Antibiotic Prophylaxis , Antiviral Agents/therapeutic use , Drug Resistance, Viral/genetics , Hematopoietic Stem Cell Transplantation , Herpes Simplex/virology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/isolation & purification , Postoperative Complications/virology , Acyclovir/pharmacology , Adolescent , Adult , Antiviral Agents/pharmacology , Child , Codon , DNA-Directed DNA Polymerase/genetics , Female , Genotype , Herpes Simplex/drug therapy , Herpes Simplex/prevention & control , Herpesvirus 1, Human/genetics , Humans , Male , Middle Aged , Molecular Sequence Data , Mutation , Phenotype , Postoperative Complications/prevention & control , Thymidine Kinase/genetics
16.
Antimicrob Agents Chemother ; 48(6): 2331-3, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155247

ABSTRACT

A colorimetric yield reduction assay, ELVIRA (enzyme-linked virus inhibitor reporter assay) HSV, was developed to determine the antiviral drug susceptibilities of herpes simplex virus (HSV). It uses an HSV-inducible reporter cell line. This simple and rapid assay has an objective readout, low inoculum size, and good reproducibility. The results correlate well with those of the plaque reduction assay.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Simplexvirus/drug effects , Acyclovir/pharmacology , Colorimetry , Foscarnet/pharmacology , Genes, Reporter/genetics , Lac Operon/genetics , Simplexvirus/genetics , Viral Plaque Assay , Virus Replication/drug effects
17.
J Clin Virol ; 30(1): 39-44, 2004 May.
Article in English | MEDLINE | ID: mdl-15072752

ABSTRACT

BACKGROUND: Detection of herpes viruses can be significantly improved by PCR. The development of real-time PCR, which has overcome several limitations of conventional PCR, improved the prospects for implementation of PCR-based assays in diagnostic laboratory. OBJECTIVES: To compare the diagnostic performance of an automated sample extraction procedure in combination with an internally controlled real-time PCR assay for detection of herpes simplex virus (HSV) and varicella-zoster virus (VZV) to conventional shell vial culture. STUDY DESIGN: One hundred eighty-two consecutive specimens from patients suspected of HSV or VZV infection were examined by internally controlled PCR and shell vial culture. An internal control consisting of phocine herpes virus was processed along with the specimens during the entire procedure and permitted to monitor extraction and amplification efficiency, including inhibition. RESULTS: A total of 48 (26.4%) specimens were positive for HSV or VZV by culture, and 77 (42.3%) by real-time PCR. Thus, overall sensitivity increased by 60.4%. All culture-positive specimens were detected and typed correctly by PCR, except for a single specimen that contained PCR inhibitors. Specifically, the real-time PCR assay increased the detection rate for HSV-1 and HSV-2 by 43.9% and 62.5%, respectively. In PCR-positive specimens, lower levels of viral DNA were found in culture-negative than in culture-positive specimens. The increase of HSV detection rates by PCR varied with the origin of specimen and was particularly significant for skin specimens (7/14 versus 3/14 detected by culture) and bronchoalveolar lavages (8/8 versus 1/8). In addition, real-time PCR significantly increased the detection rate for VZV. CONCLUSIONS: Compared to shell vial culture, our real-time PCR assay demonstrated a superior sensitivity and an added value of using internal control for checking the quality of examination of each specimen. These results provide a solid basis for implementation of real-time PCR in the routine diagnosis of HSV and VZV infections in various clinical specimens.


Subject(s)
Herpes Simplex/diagnosis , Herpes Zoster/diagnosis , Herpesvirus 1, Human/isolation & purification , Herpesvirus 2, Human/isolation & purification , Herpesvirus 3, Human/isolation & purification , Polymerase Chain Reaction/methods , Automation , Bronchoalveolar Lavage Fluid/virology , DNA, Viral/blood , Herpes Simplex/virology , Herpes Zoster/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/growth & development , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/growth & development , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/growth & development , Humans , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Skin/virology , Virus Cultivation , Virus Replication
18.
Antivir Ther ; 9(1): 97-104, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15040541

ABSTRACT

Sequential herpes simplex virus type 1 (HSV-1) isolates were obtained from a paediatric haematopoietic stem cell transplant (HSCT) patient who received prolonged therapy with acyclovir (ACV) followed by foscarnet (PFA) and topical cidofovir (HPMPC) for severe persistent mucocutaneous HSV-1 infection. The isolates were retrospectively studied for drug resistance. The first resistant isolate associated with clinical failure of antiviral therapy emerged 44 days post-ACV treatment initiation. Susceptibility testing revealed an ACV-resistant HSV strain that demonstrated cross resistance to PFA in the absence of any previous PFA treatment. The observed cross resistance was conferred by a single amino acid substitution, Ser724Asn, in the HSV DNA polymerase (DNA pol) gene. During the subsequent course of ACV therapy, the ACV/PFA-cross-resistant isolates were replaced by ACV-resistant, PFA-sensitive isolates. These isolates carried no DNA pol mutations, but had an Arg163His substitution in the thymidine kinase gene. Upon subsequent switching of antiviral therapy from ACV to PFA, the original ACV/PFA-cross-resistant DNA pol mutant re-appeared. Our study shows the emergence of different drug-resistant HSV variants during ongoing, unchanged ACV therapy. Furthermore, a rapid re-selection of the original resistant variant was observed after switch. For optimal antiviral management of HSV infections in HSCT recipients, therapeutic decisions should be guided by drug susceptibility results whenever therapeutic failure is observed and/or when changes in antiviral treatment are considered.


Subject(s)
DNA-Directed DNA Polymerase/genetics , Herpes Simplex/immunology , Herpesvirus 1, Human/drug effects , Immunocompromised Host/immunology , Thymidine Kinase/genetics , Amino Acid Substitution , Child , DNA-Directed DNA Polymerase/metabolism , Genotype , Herpesvirus 1, Human/enzymology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/isolation & purification , Humans , Infant, Newborn , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Male , Microbial Sensitivity Tests , Mouth/virology , Nose/virology , Phylogeny , Sputum/virology , Stem Cell Transplantation/adverse effects , Thymidine Kinase/metabolism
19.
Antimicrob Agents Chemother ; 46(9): 2943-7, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183251

ABSTRACT

A quantitative real-time PCR (TaqMan) assay was developed for determination of antiviral drug susceptibility of herpes simplex virus (HSV). After short-time culture of the virus, the antiviral drug susceptibility of HSV isolates for acyclovir (ACV) was determined by measuring the reduction of the HSV type 1 (HSV-1) DNA levels in culture supernatants using real-time PCR. The 50% inhibitory concentration was reported as the concentration of antiviral drug that reduced the number of HSV-1 DNA copies by 50%. A total of 15 well-characterized ACV-sensitive or -resistant strains and clinical isolates were used for assay evaluation. The new assay with real-time PCR readout permitted rapid (3 days), objective, and reproducible determination of HSV-1 drug susceptibilities with no need for stringent control of initial multiplicity of infection. Furthermore, the real-time PCR assay results showed good correlation (r = 0.86) with those for the plaque reduction assay. In conclusion, the real-time PCR assay described here is a suitable quantitative method for determination of antiviral susceptibility of HSV-1, amenable for use in the routine diagnostic virology laboratory.


Subject(s)
Antiviral Agents/pharmacology , Reverse Transcriptase Polymerase Chain Reaction/methods , Simplexvirus/drug effects , Animals , Chlorocebus aethiops , Computer Systems , DNA Replication/drug effects , DNA, Viral/analysis , DNA, Viral/biosynthesis , Herpesvirus 1, Human/drug effects , Kinetics , Reproducibility of Results , Vero Cells , Viral Plaque Assay
20.
Cancer Immunol Immunother ; 51(2): 111-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11904736

ABSTRACT

To target the E7 protein of human papilloma virus 16 to the cell surface, a fusion gene was constructed. It encodes the signal peptide, part of the immunoglobulin (IgG)-like domain, the transmembrane anchor of vaccinia virus (VV) hemagglutinin (HA), and the complete E7-coding sequence. The fusion gene was expressed under the HA late promoter by a recombinant VV, designated VV-E7-HA. The E7-HA protein was displayed on the surface of cells infected with the recombinant virus and was more stable than unmodified E7. The biological properties of the VV-E7-HA virus were compared with those of a VV-E7 virus that expressed the unmodified E7 and with a VV expressing the Sig-E7-LAMP fusion protein. While the first two of these recombinants were based on VV strain Praha, the third was derived from the WR strain of VV. Infection of mice with the VV-E7-HA virus induced the formation of E7-specific antibodies with the predominance of the IgG2a isotype, whereas the other two viruses did not induce the formation of E7-specific antibodies. Unlike the other two viruses, VV-E7-HA did not induce a response of cytotoxic T lymphocytes or Th1 cells and did not protect mice against the growth of E7-expressing tumors. Thus, VV-E7-HA induced a differently polarized immune response to the E7 protein than the other two viruses.


Subject(s)
Oncogene Proteins, Viral/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/biosynthesis , Hemagglutinins, Viral/immunology , Immunity, Cellular , Interleukin-4/biosynthesis , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/prevention & control , Oncogene Proteins, Viral/analysis , Papillomavirus E7 Proteins , Rabbits , Recombinant Fusion Proteins/immunology , Vaccination , Vaccinia virus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...