Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Ther Methods Clin Dev ; 14: 126-133, 2019 Sep 13.
Article in English | MEDLINE | ID: mdl-31338384

ABSTRACT

Preexisting immunity against adeno-associated virus (AAV) is a major challenge facing AAV gene therapy, resulting in the exclusion of patients from clinical trials. Accordingly, proper assessment of anti-AAV immunity is necessary for understanding clinical data and for product development. Previous studies on anti-AAV prevalence lack method standardization, rendering the assessment of prevalence difficult. Addressing this need, we used clinical assays that were validated according to guidelines for a comprehensive characterization of anti-AAV1, -AAV2, -AAV5, and -AAV8 immunity in large international cohorts of healthy donors and patients with hemophilia B. Here, we report a higher than expected average prevalence for anti-AAV8 (∼40%) and anti-AAV5 (∼30%) neutralizing antibodies (NAbs), which is supported by strongly correlating anti-AAV IgG antibody titers. A similar anti-AAV8 NAb prevalence was observed in hemophilia B patients. In addition, a high co-prevalence of NAbs against other serotypes makes switching to gene therapy using another serotype difficult. As anti-AAV T cell responses are believed to influence transduction, we characterized anti-AAV T cell responses using interleukin-2 (IL-2) and interferon-γ (IFN-γ) ELISpot assays, revealing a similar prevalence of IFN-γ responses (∼20%) against different serotypes that did not correlate with NAbs. These data, along with the long-term stability of NAbs, emphasize the need to develop strategies to circumvent anti-AAV immunity.

2.
Hum Gene Ther Methods ; 30(2): 35-43, 2019 04.
Article in English | MEDLINE | ID: mdl-30734588

ABSTRACT

Patients with preexisting anti-adeno-associated virus serotype 8 (AAV8) neutralizing antibodies (NAbs) are currently excluded from AAV8 gene therapy trials. Therefore, the assessment of biologically relevant AAV8-NAb titers is critical for product development in gene therapy. However, standardized assays have not been routinely used to determine anti-AAV8-NAb titers, contributing to a wide range of reported anti-AAV8 prevalence rates. Using a clinical in vitro NAb assay in a separate study, a higher than expected anti-AAV8-NAb prevalence of about 50% was found in international cohorts. This comparative study has a translational character, confirming the biological relevance of anti-AAV8-antibody titers measured by this assay. The significance of low-titer anti-AAV8 NAbs is shown, along with the relevance of the in vitro assay cutoff (1:5) compared with other assays. Importantly, internally standardized reagents and purified AAV8 constructs containing 90% full capsids were used to reduce the effect of empty capsids. It was found that even very low anti-AAV8-NAb titers (<1:5) could efficiently hinder transduction in vivo, demonstrating the importance of sensitive NAb assays for clinical applications. The in vitro NAb assay was found to be more sensitive than an in vivo NAb assay and thus more suitable for patient screening. Additionally, the study showed that anti-AAV8-NAb titers <1:5 were very rare, further supporting the in vitro assay. However, assays using a lower cutoff may still be useful to explain potential variances in transgene expression. These findings support the relevance of the higher than expected prevalence of anti-AAV8 NAbs, highlighting the need for strategies to circumvent preexisting anti-AAV8 NAbs.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dependovirus/immunology , Animals , Antibodies, Monoclonal/immunology , Biological Assay , Cell Line, Tumor , Factor IX/genetics , Factor IX/immunology , Genetic Therapy , Genetic Vectors , Humans , Mice, Transgenic , Viral Proteins/genetics , Viral Proteins/immunology
3.
J Int AIDS Soc ; 20(1): 21865, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28953327

ABSTRACT

INTRODUCTION: Restriction factors (RFs) suppress HIV-1 in cell lines and primary cell models. Hence, RFs might be attractive targets for novel antiviral strategies, but their importance for virus control in vivo is controversial. METHODS: We profiled the expression of RFs in primary blood-derived mononuclear cells (PBMC) from therapy-naïve HIV-1 patients and quantified infection. RESULTS: Overall, there was no correlation between individual RF expression and HIV-1 status in total PBMC. However, we identified a T cell population with low levels of intracellular CD2 and reduced expression of SAMHD1, p21 and SerinC5. CD2low T cells with reduced RF expression were markedly positive for HIV-1 p24. In contrast, CD2+ T cells were less infected and expressed higher levels of RFs. CD2low T cell infection correlated with viral loads and was associated with HIV-1 disease progression. CONCLUSIONS: In untreated therapy naïve chronic HIV-1 patients, RF expression in T cells is associated with CD2 expression and seems to influence viral loads. Our study suggests that RFs help to control HIV-1 infection in certain T cells in vivo and supports the potential for RFs as promising targets for therapeutic intervention.


Subject(s)
HIV Infections/genetics , HIV Infections/immunology , HIV-1/physiology , T-Lymphocytes/immunology , Adult , Aged , Female , HIV Infections/virology , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Male , Middle Aged , SAM Domain and HD Domain-Containing Protein 1/genetics , SAM Domain and HD Domain-Containing Protein 1/immunology , T-Lymphocytes/virology , Viral Load
4.
Open Biol ; 6(7)2016 07.
Article in English | MEDLINE | ID: mdl-27383627

ABSTRACT

The majority of T cells encountered by HIV-1 are non-activated and do not readily allow productive infection. HIV-1 Vpr is highly abundant in progeny virions, and induces signalling and HIV-1 LTR transcription. We hence hypothesized that Vpr might be a determinant of non-activated T-cell infection. Virion-delivered Vpr activated nuclear factor of activated T cells (NFAT) through Ca(2+) influx and interference with the NFAT export kinase GSK3ß. This leads to NFAT translocation and accumulation within the nucleus and was required for productive infection of unstimulated primary CD4(+) T cells. A mutagenesis approach revealed correlation of Vpr-mediated NFAT activation with its ability to enhance LTR transcription and mediate cell cycle arrest. Upon NFAT inhibition, Vpr did not augment resting T-cell infection, and showed reduced G2/M arrest and LTR transactivation. Altogether, Vpr renders unstimulated T cells more permissive for productive HIV-1 infection and stimulates activation of productively infected as well as virus-exposed T cells. Therefore, it could be involved in the establishment and reactivation of HIV-1 from viral reservoirs and might have an impact on the levels of immune activation, which are determinants of HIV-1 pathogenesis.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Long Terminal Repeat , NFATC Transcription Factors/metabolism , Virion/genetics , vpr Gene Products, Human Immunodeficiency Virus/metabolism , CD4-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Cell Nucleus/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , HeLa Cells , Humans , Jurkat Cells , Transcription, Genetic , Virion/metabolism , vpr Gene Products, Human Immunodeficiency Virus/genetics
5.
Sci Rep ; 6: 20394, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26833261

ABSTRACT

Novel therapeutic options are urgently needed to improve global treatment of virus infections. Herbal products with confirmed clinical safety features are attractive starting material for the identification of new antiviral activities. Here we demonstrate that Cistus incanus (Ci) herbal products inhibit human immunodeficiency virus (HIV) infections in vitro. Ci extract inhibited clinical HIV-1 and HIV-2 isolates, and, importantly, a virus isolate with multiple drug resistances, confirming broad anti-HIV activity. Antiviral activity was highly selective for virus particles, preventing primary attachment of the virus to the cell surface and viral envelope proteins from binding to heparin. Bioassay-guided fractionation indicated that Ci extract contains numerous antiviral compounds and therefore has favorably low propensity to induce virus resistance. Indeed, no resistant viruses emerged during 24 weeks of continuous propagation of the virus in the presence of Ci extracts. Finally, Ci extracts also inhibited infection by virus particles pseudotyped with Ebola and Marburg virus envelope proteins, indicating that antiviral activity of Ci extract extends to emerging viral pathogens. These results demonstrate that Ci extracts show potent and broad in vitro antiviral activity against viruses that cause life-threatening diseases in humans and are promising sources of agents that target virus particles.


Subject(s)
Antiviral Agents/pharmacology , Cistus/chemistry , Filoviridae/drug effects , HIV-1/drug effects , Plant Extracts/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Drug Resistance, Viral , Humans , Microbial Sensitivity Tests , Plant Extracts/chemistry , Polyphenols/chemistry , Polyphenols/pharmacology , Virus Replication/drug effects
6.
Mol Cell Proteomics ; 14(8): 2085-102, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25991687

ABSTRACT

Naive CD4(+) T cells are the common precursors of multiple effector and memory T-cell subsets and possess a high plasticity in terms of differentiation potential. This stem-cell-like character is important for cell therapies aiming at regeneration of specific immunity. Cell surface proteins are crucial for recognition and response to signals mediated by other cells or environmental changes. Knowledge of cell surface proteins of human naive CD4(+) T cells and their changes during the early phase of T-cell activation is urgently needed for a guided differentiation of naive T cells and may support the selection of pluripotent cells for cell therapy. Periodate oxidation and aniline-catalyzed oxime ligation technology was applied with subsequent quantitative liquid chromatography-tandem MS to generate a data set describing the surface proteome of primary human naive CD4(+) T cells and to monitor dynamic changes during the early phase of activation. This led to the identification of 173 N-glycosylated surface proteins. To independently confirm the proteomic data set and to analyze the cell surface by an alternative technique a systematic phenotypic expression analysis of surface antigens via flow cytometry was performed. This screening expanded the previous data set, resulting in 229 surface proteins, which were expressed on naive unstimulated and activated CD4(+) T cells. Furthermore, we generated a surface expression atlas based on transcriptome data, experimental annotation, and predicted subcellular localization, and correlated the proteomics result with this transcriptional data set. This extensive surface atlas provides an overall naive CD4(+) T cell surface resource and will enable future studies aiming at a deeper understanding of mechanisms of T-cell biology allowing the identification of novel immune targets usable for the development of therapeutic treatments.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Lymphocyte Activation/immunology , Proteomics/methods , Receptors, Antigen, T-Cell/metabolism , Cell Membrane/metabolism , Cluster Analysis , Computer Simulation , Flow Cytometry , Gene Expression Profiling , Gene Ontology , Glycoproteins/metabolism , Humans , Proteome/metabolism , Reproducibility of Results , Transcriptome/genetics
7.
J Virol ; 89(10): 5687-700, 2015 May.
Article in English | MEDLINE | ID: mdl-25822027

ABSTRACT

UNLABELLED: Leukocyte recirculation between blood and lymphoid tissues is required for the generation and maintenance of immune responses against pathogens and is crucially controlled by the L-selectin (CD62L) leukocyte homing receptor. CD62L has adhesion and signaling functions and initiates the capture and rolling on the vascular endothelium of cells entering peripheral lymph nodes. This study reveals that CD62L is strongly downregulated on primary CD4(+) T lymphocytes upon infection with human immunodeficiency virus type 1 (HIV-1). Reduced cell surface CD62L expression was attributable to the Nef and Vpu viral proteins and not due to increased shedding via matrix metalloproteases. Both Nef and Vpu associated with and sequestered CD62L in perinuclear compartments, thereby impeding CD62L transport to the plasma membrane. In addition, Nef decreased total CD62L protein levels. Importantly, infection with wild-type, but not Nef- and Vpu-deficient, HIV-1 inhibited the capacity of primary CD4(+) T lymphocytes to adhere to immobilized fibronectin in response to CD62L ligation. Moreover, HIV-1 infection impaired the signaling pathways and costimulatory signals triggered in primary CD4(+) T cells by CD62L ligation. We propose that HIV-1 dysregulates CD62L expression to interfere with the trafficking and activation of infected T cells. Altogether, this novel HIV-1 function could contribute to virus dissemination and evasion of host immune responses. IMPORTANCE: L-selectin (CD62L) is an adhesion molecule that mediates the first steps of leukocyte homing to peripheral lymph nodes, thus crucially controlling the initiation and maintenance of immune responses to pathogens. Here, we report that CD62L is downmodulated on the surfaces of HIV-1-infected T cells through the activities of two viral proteins, Nef and Vpu, that prevent newly synthesized CD62L molecules from reaching the plasma membrane. We provide evidence that CD62L downregulation on HIV-1-infected primary T cells results in impaired adhesion and signaling functions upon CD62L triggering. Removal of cell surface CD62L may predictably keep HIV-1-infected cells away from lymph nodes, the privileged sites of both viral replication and immune response activation, with important consequences, such as systemic viral spread and evasion of host immune surveillance. Altogether, we propose that Nef- and Vpu-mediated subversion of CD62L function could represent a novel determinant of HIV-1 pathogenesis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV Infections/immunology , HIV Infections/virology , HIV-1/immunology , HIV-1/pathogenicity , Human Immunodeficiency Virus Proteins/immunology , L-Selectin/metabolism , Viral Regulatory and Accessory Proteins/immunology , nef Gene Products, Human Immunodeficiency Virus/immunology , Cell Adhesion/immunology , Cells, Cultured , Down-Regulation , HIV-1/physiology , Host-Pathogen Interactions/immunology , Humans , Jurkat Cells , Lymphocyte Activation , Protein Transport , Signal Transduction/immunology
8.
J Virol ; 89(6): 3247-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25568205

ABSTRACT

UNLABELLED: Tetraspanins constitute a family of cellular proteins that organize various membrane-based processes. Several members of this family, including CD81, are actively recruited by HIV-1 Gag to viral assembly and release sites. Despite their enrichment at viral exit sites, the overall levels of tetraspanins are decreased in HIV-1-infected cells. Here, we identify Vpu as the main viral determinant for tetraspanin downregulation. We also show that reduction of CD81 levels by Vpu is not a by-product of CD4 or BST-2/tetherin elimination from the surfaces of infected cells and likely occurs through an interaction between Vpu and CD81. Finally, we document that Vpu-mediated downregulation of CD81 from the surfaces of infected T cells can contribute to preserving the infectiousness of viral particles, thus revealing a novel Vpu function that promotes virus propagation by modulating the host cell environment. IMPORTANCE: The HIV-1 accessory protein Vpu has previously been shown to downregulate various host cell factors, thus helping the virus to overcome restriction barriers, evade immune attack, and maintain the infectivity of viral particles. Our study identifies tetraspanins as an additional group of host factors whose expression at the surfaces of infected cells is lowered by Vpu. While the downregulation of these integral membrane proteins, including CD81 and CD82, likely affects more than one function of HIV-1-infected cells, we document that Vpu-mediated lowering of CD81 levels in viral particles can be critical to maintaining their infectiousness.


Subject(s)
Down-Regulation , HIV Infections/genetics , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/metabolism , Tetraspanin 28/genetics , Viral Regulatory and Accessory Proteins/metabolism , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , Host-Pathogen Interactions , Human Immunodeficiency Virus Proteins/genetics , Humans , Protein Binding , Tetraspanin 28/metabolism , Viral Regulatory and Accessory Proteins/genetics
9.
PLoS One ; 9(8): e105478, 2014.
Article in English | MEDLINE | ID: mdl-25127017

ABSTRACT

The TAR DNA binding protein (TDP-43) was originally identified as a host cell factor binding to the HIV-1 LTR and thereby suppressing HIV-1 transcription and gene expression (Ou et al., J.Virol. 1995, 69(6):3584). TDP-43 is a global regulator of transcription, can influence RNA metabolism in many different ways and is ubiquitously expressed. Thus, TDP-43 could be a major factor restricting HIV-1 replication at the level of LTR transcription and gene expression. These facts prompted us to revisit the role of TDP-43 for HIV-1 replication. We utilized established HIV-1 cell culture systems as well as primary cell models and performed a comprehensive analysis of TDP-43 function and investigated its putative impact on HIV-1 gene expression. In HIV-1 infected cells TDP-43 was neither degraded nor sequestered from the nucleus. Furthermore, TDP-43 overexpression as well as siRNA mediated knockdown did not affect HIV-1 gene expression and virus production in T cells and macrophages. In summary, our experiments argue against a restricting role of TDP-43 during HIV-1 replication in immune cells.


Subject(s)
DNA-Binding Proteins/metabolism , HIV-1/physiology , Virus Replication , DNA-Binding Proteins/genetics , Gene Expression , Gene Expression Regulation, Viral , Genes, Viral , HEK293 Cells , HIV Long Terminal Repeat , Humans , Jurkat Cells , Protein Transport , Transcriptional Activation
10.
Science ; 343(6176): 1221-8, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24557838

ABSTRACT

Current antiviral agents can control but not eliminate hepatitis B virus (HBV), because HBV establishes a stable nuclear covalently closed circular DNA (cccDNA). Interferon-α treatment can clear HBV but is limited by systemic side effects. We describe how interferon-α can induce specific degradation of the nuclear viral DNA without hepatotoxicity and propose lymphotoxin-ß receptor activation as a therapeutic alternative. Interferon-α and lymphotoxin-ß receptor activation up-regulated APOBEC3A and APOBEC3B cytidine deaminases, respectively, in HBV-infected cells, primary hepatocytes, and human liver needle biopsies. HBV core protein mediated the interaction with nuclear cccDNA, resulting in cytidine deamination, apurinic/apyrimidinic site formation, and finally cccDNA degradation that prevented HBV reactivation. Genomic DNA was not affected. Thus, inducing nuclear deaminases-for example, by lymphotoxin-ß receptor activation-allows the development of new therapeutics that, in combination with existing antivirals, may cure hepatitis B.


Subject(s)
Antiviral Agents/pharmacology , DNA, Circular/metabolism , DNA, Viral/metabolism , Hepatitis B virus/drug effects , Hepatitis B/drug therapy , Hepatocytes/drug effects , Interferon-alpha/pharmacology , Lymphotoxin beta Receptor/agonists , Animals , Antibodies, Monoclonal , Antiviral Agents/therapeutic use , Cell Line , Cell Nucleus/virology , Cytidine/metabolism , Cytidine Deaminase/biosynthesis , Hepatitis B virus/metabolism , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Interferon-alpha/therapeutic use , Liver/drug effects , Liver/metabolism , Liver/virology , Lymphotoxin beta Receptor/antagonists & inhibitors , Mice, SCID , Minor Histocompatibility Antigens , Proteins , Up-Regulation
11.
PLoS One ; 9(1): e87487, 2014.
Article in English | MEDLINE | ID: mdl-24489923

ABSTRACT

Global HIV-1 treatment would benefit greatly from safe herbal medicines with scientifically validated novel anti-HIV-1 activities. The root extract from the medicinal plant Pelargonium sidoides (PS) is licensed in Germany as the herbal medicine EPs®7630, with numerous clinical trials supporting its safety in humans. Here we provide evidence from multiple cell culture experiments that PS extract displays potent anti-HIV-1 activity. We show that PS extract protects peripheral blood mononuclear cells and macrophages from infection with various X4 and R5 tropic HIV-1 strains, including clinical isolates. Functional studies revealed that the extract from PS has a novel mode-of-action. It interferes directly with viral infectivity and blocks the attachment of HIV-1 particles to target cells, protecting them from virus entry. Analysis of the chemical footprint of anti-HIV activity indicates that HIV-1 inhibition is mediated by multiple polyphenolic compounds with low cytotoxicity and can be separated from other extract components with higher cytotoxicity. Based on our data and its excellent safety profile, we propose that PS extract represents a lead candidate for the development of a scientifically validated herbal medicine for anti-HIV-1 therapy with a mode-of-action different from and complementary to current single-molecule drugs.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Pelargonium/chemistry , Plant Extracts/pharmacology , Plant Roots/chemistry , Virus Attachment/drug effects , Anti-HIV Agents/chemistry , Anti-HIV Agents/isolation & purification , Drug Evaluation, Preclinical , HEK293 Cells , HIV Infections/drug therapy , Humans , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plants, Medicinal/chemistry , Polyphenols/chemistry , Polyphenols/isolation & purification , Polyphenols/pharmacology
12.
Retrovirology ; 11: 1, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24383984

ABSTRACT

BACKGROUND: Increased cellular iron levels are associated with high mortality in HIV-1 infection. Moreover iron is an important cofactor for viral replication, raising the question whether highly divergent lentiviruses actively modulate iron homeostasis. Here, we evaluated the effect on cellular iron uptake upon expression of the accessory protein Nef from different lentiviral strains. RESULTS: Surface Transferrin receptor (TfR) levels are unaffected by Nef proteins of HIV-1 and its simian precursors but elevated in cells expressing Nefs from most other primate lentiviruses due to reduced TfR internalization. The SIV Nef-mediated reduction of TfR endocytosis is dependent on an N-terminal AP2 binding motif that is not required for downmodulation of CD4, CD28, CD3 or MHCI. Importantly, SIV Nef-induced inhibition of TfR endocytosis leads to the reduction of Transferrin uptake and intracellular iron concentration and is accompanied by attenuated lentiviral replication in macrophages. CONCLUSION: Inhibition of Transferrin and thereby iron uptake by SIV Nef might limit viral replication in myeloid cells. Furthermore, this new SIV Nef function could represent a virus-host adaptation that evolved in natural SIV-infected monkeys.


Subject(s)
Endocytosis/drug effects , Gene Products, nef/metabolism , Iron/metabolism , Receptors, Transferrin/antagonists & inhibitors , Simian Immunodeficiency Virus/physiology , Transferrin/metabolism , Animals , Haplorhini
13.
Retrovirology ; 9: 82, 2012 Oct 04.
Article in English | MEDLINE | ID: mdl-23035819

ABSTRACT

Macrophages are important target cells for the Human Immunodeficiency Virus Type I (HIV-1) in vivo. Several studies have assessed the molecular biology of the virus in this cell type, and a number of differences towards HIV-1 infection of CD4+ T cells have been described. There is a broad consensus that macrophages resist HIV-1 infection much better than CD4+ T cells. Among other reasons, this is due to the presence of the recently identified host cell restriction factor SamHD1, which is strongly expressed in cells of the myeloid lineage. Furthermore, macrophages produce and release relatively low amounts of infectious HIV-1 and are less sensitive to viral cytotoxicity in comparison to CD4+ T cells. Nevertheless, macrophages play a crucial role in the different phases of HIV-1 infection. In this review, we summarize and discuss the significance of macrophages for HIV-1 transmission, the acute and chronic phases of HIV-1 infection, the development of acquired immunodeficiency syndrome (AIDS) and HIV-associated diseases, including neurocognitive disorders. We propose that interaction of HIV-1 with macrophages is crucial during all stages of HIV-1 infection. Thus, long-term successful treatment of HIV-1 infected individuals requires potent strategies to prevent HIV-1 from entering and persisting in these cells.


Subject(s)
HIV-1/immunology , HIV-1/pathogenicity , Macrophages/immunology , Macrophages/virology , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/pathology , Host-Pathogen Interactions , Humans
14.
Cancer Lett ; 319(2): 164-172, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22261339

ABSTRACT

To better understand the response of HCC to EGFR inhibition, we analyzed factors connected to the resistance of HCC cells against gefitinib. Sensitive HCC3 cells co-expressed EGFR and ErbB3 but lacked kinase-domain mutations in EGFR. Interestingly, expression of MVP was restricted to resistant cell lines, whereas ABCB1 and ABCC1 showed no association with gefitinib resistance. Moreover, ectopic MVP expression in HCC3 cells decreased gefitinib sensitivity, increased AKT phosphorylation and reduced the expression of inflammatory pathway-associated genes, whereas silencing of MVP in Hep3B and HepG2 cells increased sensitivity. These findings suggest MVP as a novel player in resistance against EGFR inhibition.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm/drug effects , ErbB Receptors/antagonists & inhibitors , Liver Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Vault Ribonucleoprotein Particles/physiology , Cell Line, Tumor , Cytokines , ErbB Receptors/genetics , Gefitinib , Humans , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects
15.
J Virol ; 86(5): 2826-36, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22205742

ABSTRACT

In macrophages, HIV-1 accumulates in intracellular vesicles designated virus-containing compartments (VCCs). These might play an important role in the constitution of macrophages as viral reservoirs and allow HIV-1 to evade the immune system by sequestration in an internal niche, which is difficult to access from the exterior. However, until now, evidence of whether internal virus accumulations are protected from the host's humoral immune response is still lacking. In order to be able to study the formation and antibody accessibility of VCCs, we generated HIV-1 with green fluorescent protein (GFP)-tagged Gag replicating in primary macrophages. Live-cell observations revealed faint initial cytosolic Gag expression and subsequent large intracellular Gag accumulations which stayed stable over days. Taking advantage of the opportunity to study the accessibility of intracellular VCCs via the cell surface, we demonstrate that macrophage internal HIV-1-containing compartments cannot be targeted by neutralizing antibodies. Furthermore, HIV-1 was efficiently transferred from antibody-treated macrophages to T cells. Three-dimensional reconstruction of electron microscopic slices revealed that Gag accumulations correspond to viral particles within enclosed compartments and convoluted membranes. Thus, although some VCCs were connected to the plasma membrane, the complex membrane architecture of the HIV-1-containing compartment might shield viral particles from neutralizing antibodies. In sum, our study provides evidence that HIV-1 is sequestered into a macrophage internal membranous web, posing an obstacle for the elimination of this viral reservoir.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , HIV Infections/immunology , HIV-1/immunology , Macrophages/immunology , Cell Line , Gene Expression Regulation, Viral , HIV Infections/virology , HIV-1/physiology , Humans , Immune Evasion , Macrophages/virology , Virus Replication
16.
Virology ; 416(1-2): 75-85, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21601230

ABSTRACT

While the C-terminal domain of HIV-1 Vpu is critical for CD4 degradation, the transmembrane domain (TM) mediates ion channel activity, enhances virus release and is essential for counteracting CD317/Bst-2/Tetherin. Here we analyzed whether the ion channel activity of Vpu is required to antagonize CD317-mediated restriction of virion release. We examined TM-mutants of three conserved residues: the S23A mutation, which was previously shown to abrogate ion channel function, did not affect Vpu mediated augmentation of virus release. In contrast, the A14N and A18N mutation did not affect ion channel activity of Vpu, but substantially reduced its ability to support virus release and to down-regulate CD317 from the cell surface. Altogether, our data suggest that not the ion channel activity of Vpu, but its ability to remove CD317 from the cell surface is required to augment HIV-1 release.


Subject(s)
Antigens, CD/metabolism , Gene Expression Regulation, Viral/physiology , HIV-1/metabolism , Human Immunodeficiency Virus Proteins/metabolism , Ion Channels/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Cell Line , Down-Regulation , GPI-Linked Proteins/metabolism , Human Immunodeficiency Virus Proteins/genetics , Humans , Viral Regulatory and Accessory Proteins/genetics , Virus Release
17.
PLoS One ; 5(2): e9344, 2010 Feb 22.
Article in English | MEDLINE | ID: mdl-20179761

ABSTRACT

BACKGROUND: Försters resonance energy transfer (FRET) microscopy is widely used for the analysis of protein interactions in intact cells. However, FRET microscopy is technically challenging and does not allow assessing interactions in large cell numbers. To overcome these limitations we developed a flow cytometry-based FRET assay and analysed interactions of human and simian immunodeficiency virus (HIV and SIV) Nef and Vpu proteins with cellular factors, as well as HIV Rev multimer-formation. RESULTS: Amongst others, we characterize the interaction of Vpu with CD317 (also termed Bst-2 or tetherin), a host restriction factor that inhibits HIV release from infected cells and demonstrate that the direct binding of both is mediated by the Vpu membrane-spanning region. Furthermore, we adapted our assay to allow the identification of novel protein interaction partners in a high-throughput format. CONCLUSION: The presented combination of FRET and FACS offers the precious possibility to discover and define protein interactions in living cells and is expected to contribute to the identification of novel therapeutic targets for treatment of human diseases.


Subject(s)
Flow Cytometry/methods , Fluorescence Resonance Energy Transfer/methods , Protein Interaction Mapping/methods , Viral Proteins/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Binding Sites/genetics , Cell Line , GPI-Linked Proteins , Gene Products, nef/genetics , Gene Products, nef/metabolism , HIV-1/metabolism , HeLa Cells , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Humans , Immunoprecipitation , Jurkat Cells , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microscopy, Confocal , Mutation , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Simian Immunodeficiency Virus/metabolism , Transfection , Viral Proteins/genetics , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
18.
Retrovirology ; 7: 1, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20078884

ABSTRACT

BACKGROUND: The human immunodeficiency virus type 1 (HIV-1) Vpu protein degrades CD4 and counteracts a restriction factor termed tetherin (CD317; Bst-2) to enhance virion release. It has been suggested that both functions can be genetically separated by mutation of a serine residue at position 52. However, recent data suggest that the S52 phosphorylation site is also important for the ability of Vpu to counteract tetherin. To clarify this issue, we performed a comprehensive analysis of HIV-1 with a mutated casein kinase-II phosphorylation site in Vpu in various cell lines, primary blood lymphocytes (PBL), monocyte-derived macrophages (MDM) and ex vivo human lymphoid tissue (HLT). RESULTS: We show that mutation of serine 52 to alanine (S52A) entirely disrupts Vpu-mediated degradation of CD4 and strongly impairs its ability to antagonize tetherin. Furthermore, casein-kinase II inhibitors blocked the ability of Vpu to degrade tetherin. Overall, Vpu S52A could only overcome low levels of tetherin, and its activity decreased in a manner dependent on the amount of transiently or endogenously expressed tetherin. As a consequence, the S52A Vpu mutant virus was unable to replicate in macrophages, which express high levels of this restriction factor. In contrast, HIV-1 Vpu S52A caused CD4+ T-cell depletion and spread efficiently in ex vivo human lymphoid tissue and PBL, most likely because these cells express comparably low levels of tetherin. CONCLUSION: Our data explain why the effect of the S52A mutation in Vpu on virus release is cell-type dependent and suggest that a reduced ability of Vpu to counteract tetherin impairs HIV-1 replication in macrophages, but not in tissue CD4+ T cells.


Subject(s)
HIV-1/physiology , Human Immunodeficiency Virus Proteins/physiology , Macrophages/virology , Membrane Glycoproteins/antagonists & inhibitors , T-Lymphocytes/virology , Viral Regulatory and Accessory Proteins/physiology , Virus Release , Virus Replication , Amino Acid Substitution , Antigens, CD , CD4 Antigens/metabolism , Cell Line , Cells, Cultured , GPI-Linked Proteins , Human Immunodeficiency Virus Proteins/genetics , Humans , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/physiology , Viral Regulatory and Accessory Proteins/genetics
19.
Anal Biochem ; 380(1): 146-8, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18541133

ABSTRACT

Baculoviruses are widely used for protein production in insect cells, and their potential for gene transfer to mammalian cells is increasingly being recognized. Here we describe a baculovirus vector with a bicistronic mammalian expression cassette and demonstrate its suitability for efficient transient and stable protein expression in human glioblastoma cells. Bicistronic baculovirus vectors are safe, cost efficient, and easy to produce; thus, they represent an excellent gene transfer system for mammalian cells.


Subject(s)
Baculoviridae/genetics , Gene Expression Regulation , Genetic Vectors/genetics , Proteins/genetics , Animals , Cell Line, Tumor , Gene Transfer Techniques , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Time Factors , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...