Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Emerg Microbes Infect ; 13(1): 2348508, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38661085

ABSTRACT

The Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne bunyavirus that causes high mortality in humans. This enveloped virus harbors two surface glycoproteins (GP), Gn and Gc, that are released by processing of a glycoprotein precursor complex whose maturation takes place in the ER and is completed through the secretion pathway. Here, we characterized the trafficking network exploited by CCHFV GPs during viral assembly, envelopment, and/or egress. We identified membrane trafficking motifs in the cytoplasmic domains (CD) of CCHFV GPs and addressed how they impact these late stages of the viral life cycle using infection and biochemical assays, and confocal microscopy in virus-producing cells. We found that several of the identified CD motifs modulate GP transport through the retrograde trafficking network, impacting envelopment and secretion of infectious particles. Finally, we identified PACS-2 as a crucial host factor contributing to CCHFV GPs trafficking required for assembly and release of viral particles.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo , Protein Transport , Virus Assembly , Humans , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Animals , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Protein Domains , Amino Acid Motifs , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Chlorocebus aethiops , HEK293 Cells , Vero Cells
2.
J Hepatol ; 78(5): 958-970, 2023 05.
Article in English | MEDLINE | ID: mdl-36702177

ABSTRACT

BACKGROUND & AIMS: Chronic coinfection with HBV and HDV leads to the most aggressive form of chronic viral hepatitis. Herein, we aimed to elucidate the molecular mechanisms underlying the widely reported observation that HDV interferes with HBV in most coinfected patients. METHODS: Patient liver tissues, primary human hepatocytes, HepaRG cells and human liver chimeric mice were used to analyze the effect of HDV on HBV using virological and RNA-sequencing analyses, as well as RNA synthesis, stability and association assays. RESULTS: Transcriptomic analyses in cell culture and mouse models of coinfection enabled us to define an HDV-induced signature, mainly composed of interferon (IFN)-stimulated genes (ISGs). We also provide evidence that ISGs are upregulated in chronically HDV/HBV-coinfected patients but not in cells that only express HDV antigen (HDAg). Inhibition of the hepatocyte IFN response partially rescued the levels of HBV parameters. We observed less HBV RNA synthesis upon HDV infection or HDV protein expression. Additionally, HDV infection or expression of HDAg alone specifically accelerated the decay of HBV RNA, and HDAg was associated with HBV RNAs. On the contrary, HDAg expression did not affect other viruses such as HCV or SARS-CoV-2. CONCLUSIONS: Our data indicate that HDV interferes with HBV through both IFN-dependent and IFN-independent mechanisms. Specifically, we uncover a new viral interference mechanism in which proteins of a satellite virus affect the RNA production of its helper virus. Exploiting these findings could pave the way to the development of new therapeutic strategies against HBV. IMPACT AND IMPLICATIONS: Although the molecular mechanisms remained unexplored, it has long been known that despite its dependency, HDV decreases HBV viremia in patients. Herein, using in vitro and in vivo models, we showed that HDV interferes with HBV through both IFN-dependent and IFN-independent mechanisms affecting HBV RNA metabolism, and we defined the HDV-induced modulation signature. The mechanisms we uncovered could pave the way for the development of new therapeutic strategies against HBV by mimicking and/or increasing the effect of HDAg on HBV RNA. Additionally, the HDV-induced modulation signature could potentially be correlated with responsiveness to IFN-α treatment, thereby helping to guide management of HBV/HDV-coinfected patients.


Subject(s)
COVID-19 , Coinfection , Hepatitis B , Hepatitis D , Humans , Mice , Animals , Hepatitis Delta Virus/physiology , Hepatitis B virus/physiology , Interferons , Hepatitis delta Antigens/metabolism , Hepatitis D/complications , Hepatitis B/complications , Virus Replication/physiology , COVID-19/complications , SARS-CoV-2/genetics , RNA, Viral/genetics
3.
Elife ; 102021 06 30.
Article in English | MEDLINE | ID: mdl-34190687

ABSTRACT

Cell entry of enveloped viruses relies on the fusion between the viral and plasma or endosomal membranes, through a mechanism that is triggered by a cellular signal. Here we used a combination of computational and experimental approaches to unravel the main determinants of hepatitis B virus (HBV) membrane fusion process. We discovered that ERp57 is a host factor critically involved in triggering HBV fusion and infection. Then, through modeling approaches, we uncovered a putative allosteric cross-strand disulfide (CSD) bond in the HBV S glycoprotein and we demonstrate that its stabilization could prevent membrane fusion. Finally, we identified and characterized a potential fusion peptide in the preS1 domain of the HBV L glycoprotein. These results underscore a membrane fusion mechanism that could be triggered by ERp57, allowing a thiol/disulfide exchange reaction to occur and regulate isomerization of a critical CSD, which ultimately leads to the exposition of the fusion peptide.


Subject(s)
Hepatitis B Surface Antigens/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Precursors/metabolism , Viral Envelope Proteins/metabolism , Virus Attachment , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Female , Gene Expression Regulation, Viral , Hepatitis B virus , Hepatocytes , Humans , Male , Membrane Fusion , Mice , Protein Disulfide-Isomerases/genetics , Viral Envelope Proteins/genetics
5.
Nat Commun ; 10(1): 2098, 2019 05 08.
Article in English | MEDLINE | ID: mdl-31068585

ABSTRACT

Hepatitis D virus (HDV) doesn't encode envelope proteins for packaging of its ribonucleoprotein (RNP) and typically relies on the surface glycoproteins (GPs) from hepatitis B virus (HBV) for virion assembly, envelopment and cellular transmission. HDV RNA genome can efficiently replicate in different tissues and species, raising the possibility that it evolved, and/or is still able to transmit, independently of HBV. Here we show that alternative, HBV-unrelated viruses can act as helper viruses for HDV. In vitro, envelope GPs from several virus genera, including vesiculovirus, flavivirus and hepacivirus, can package HDV RNPs, allowing efficient egress of HDV particles in the extracellular milieu of co-infected cells and subsequent entry into cells expressing the relevant receptors. Furthermore, HCV can propagate HDV infection in the liver of co-infected humanized mice for several months. Further work is necessary to evaluate whether HDV is currently transmitted by HBV-unrelated viruses in humans.


Subject(s)
Coinfection/transmission , Hepatitis D/transmission , Hepatitis Delta Virus/physiology , Virus Assembly , Animals , Cell Line, Tumor , Coinfection/virology , Flavivirus/metabolism , Hepacivirus/metabolism , Hepacivirus/pathogenicity , Hepatitis D/virology , Hepatitis Delta Virus/isolation & purification , Hepatitis Delta Virus/pathogenicity , Hepatocytes/transplantation , Hepatocytes/virology , Humans , Mice , Mice, Inbred NOD , Mice, Transgenic , Primary Cell Culture , RNA, Viral/isolation & purification , Ribonucleoproteins/metabolism , Vesiculovirus/metabolism , Viral Envelope Proteins/metabolism , Virion/metabolism
6.
Blood Adv ; 3(3): 461-475, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30755435

ABSTRACT

T cells represent a valuable tool for treating cancers and infectious and inherited diseases; however, they are mainly short-lived in vivo. T-cell therapies would strongly benefit from gene transfer into long-lived persisting naive T cells or T-cell progenitors. Here we demonstrate that baboon envelope glycoprotein pseudotyped lentiviral vectors (BaEV-LVs) far outperformed other LV pseudotypes for transduction of naive adult and fetal interleukin-7-stimulated T cells. Remarkably, BaEV-LVs efficiently transduced thymocytes and T-cell progenitors generated by culture of CD34+ cells on Delta-like ligand 4 (Dll4). Upon NOD/SCIDγC-/- engraftment, high transduction levels (80%-90%) were maintained in all T-cell subpopulations. Moreover, T-cell lineage reconstitution was accelerated in NOD/SCIDγC-/- recipients after T-cell progenitor injection compared with hematopoietic stem cell transplantation. Furthermore, γC-encoding BaEV-LVs very efficiently transduced Dll4-generated T-cell precursors from a patient with X-linked severe combined immunodeficiency (SCID-X1), which fully rescued T-cell development in vitro. These results indicate that BaEV-LVs are valuable tools for the genetic modification of naive T cells, which are important targets for gene therapy. Moreover, they allowed for the generation of gene-corrected T-cell progenitors that rescued SCID-X1 T-cell development in vitro. Ultimately, the coinjection of LV-corrected T-cell progenitors and hematopoietic stem cells might accelerate T-cell reconstitution in immunodeficient patients.


Subject(s)
Lentivirus/genetics , Stem Cells/metabolism , Animals , Mice , Mice, Inbred NOD , Mice, SCID , Papio
7.
PLoS Pathog ; 13(12): e1006774, 2017 12.
Article in English | MEDLINE | ID: mdl-29253880

ABSTRACT

Viroporins are small transmembrane proteins with ion channel activities modulating properties of intracellular membranes that have diverse proviral functions. Hepatitis C virus (HCV) encodes a viroporin, p7, acting during assembly, envelopment and secretion of viral particles (VP). HCV p7 is released from the viral polyprotein through cleavage at E2-p7 and p7-NS2 junctions by signal peptidase, but also exists as an E2p7 precursor, of poorly defined properties. Here, we found that ectopic p7 expression in HCVcc-infected cells reduced secretion of particle-associated E2 glycoproteins. Using biochemical assays, we show that p7 dose-dependently slows down the ER-to-Golgi traffic, leading to intracellular retention of E2, which suggested that timely E2p7 cleavage and p7 liberation are critical events to control E2 levels. By studying HCV mutants with accelerated E2p7 processing, we demonstrate that E2p7 cleavage controls E2 intracellular expression and secretion levels of nucleocapsid-free subviral particles and infectious virions. In addition, our imaging data reveal that, following p7 liberation, the amino-terminus of p7 is exposed towards the cytosol and coordinates the encounter between NS5A and NS2-based assembly sites loaded with E1E2 glycoproteins, which subsequently leads to nucleocapsid envelopment. We identify punctual mutants at p7 membrane interface that, by abrogating NS2/NS5A interaction, are defective for transmission of infectivity owing to decreased secretion of core and RNA and to increased secretion of non/partially-enveloped particles. Altogether, our results indicate that the retarded E2p7 precursor cleavage is essential to regulate the intracellular and secreted levels of E2 through p7-mediated modulation of the cell secretory pathway and to unmask critical novel assembly functions located at p7 amino-terminus.


Subject(s)
Hepacivirus/physiology , Hepacivirus/pathogenicity , Viral Envelope Proteins/physiology , Viral Proteins/physiology , Amino Acid Sequence , Cell Line , HEK293 Cells , Hepacivirus/genetics , Hepatitis C/etiology , Hepatitis C/virology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Models, Biological , Mutation , Protein Processing, Post-Translational , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Nonstructural Proteins/physiology , Viral Proteins/chemistry , Viral Proteins/genetics , Virulence/genetics , Virulence/physiology , Virus Assembly/genetics , Virus Assembly/physiology
8.
Hepatology ; 66(6): 1750-1765, 2017 12.
Article in English | MEDLINE | ID: mdl-28445592

ABSTRACT

Chronic hepatitis B virus (HBV) infection is a major risk factor for hepatocellular carcinoma (HCC) and current treatments for chronic hepatitis B and HCC are suboptimal. Herein, we identified cellular serine/threonine Polo-like-kinase 1 (PLK1) as a positive effector of HBV replication. The aim of this study was to demonstrate the proviral role of PLK1 in HBV biosynthesis and validate PLK1 inhibition a potential antiviral strategy. To this end, we employed physiologically relevant HBV infection models of primary human hepatocytes (PHHs) and differentiated HepaRG cells in conjunction with pharmacologic PLK1 inhibitors, small interfering RNA (siRNA)-mediated knockdown, and overexpression of constitutively active PLK1 (PLK1CA ). In addition, a humanized liver Fah-/- /Rag2-/- /Il2rg-/- (FRG) mouse model was used to determine the antiviral effect of PLK1 inhibitor BI-2536 on HBV infection in vivo. Finally, in vitro PLK1 kinase assays and site-directed mutagenesis were employed to demonstrate that HBV core protein (HBc) is a PLK1 substrate. We demonstrated that HBV infection activated cellular PLK1 in PHHs and differentiated HepaRG cells. PLK1 inhibition by BI-2536 or siRNA-mediated knockdown suppressed HBV DNA biosynthesis, whereas overexpression of PLK1CA increased it, suggesting that the PLK1 effects on viral biosynthesis are specific and that PLK1 is a proviral cellular factor. Significantly, BI-2536 administration to HBV-infected humanized liver FRG mice strongly inhibited HBV infection, validating PLK1 as an antiviral target in vivo. The proviral action of PLK1 is associated with the biogenesis of the nucleocapsid, as BI-2536 leads to its decreased intracellular formation/accumulation. In this respect, our studies identified HBc as a PLK1 substrate in vitro, and mapped PLK1 phosphorylation sites on this protein. CONCLUSION: PLK1 is a proviral host factor that could be envisaged as a target for combined antiviral and antitumoral strategies against HBV infection and HBV-mediated carcinogenesis. (Hepatology 2017;66:1750-1765).


Subject(s)
Cell Cycle Proteins/metabolism , Hepatitis B virus/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Pteridines/therapeutic use , Viral Core Proteins/metabolism , Virus Replication , Amino Acid Sequence , Animals , Cell Cycle Proteins/antagonists & inhibitors , Drug Evaluation, Preclinical , Enzyme Activation , Hepatocytes/enzymology , Host-Pathogen Interactions , Humans , Mice , Phosphorylation , Primary Cell Culture , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Polo-Like Kinase 1
9.
Blood Adv ; 1(23): 2088-2104, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29296856

ABSTRACT

Hematopoietic stem cell (HSC)-based gene therapy trials are now moving toward the use of lentiviral vectors (LVs) with success. However, one challenge in the field remains: efficient transduction of HSCs without compromising their stem cell potential. Here we showed that measles virus glycoprotein-displaying LVs (hemagglutinin and fusion protein LVs [H/F-LVs]) were capable of transducing 100% of early-acting cytokine-stimulated human CD34+ (hCD34+) progenitor cells upon a single application. Strikingly, these H/F-LVs also allowed transduction of up to 70% of nonstimulated quiescent hCD34+ cells, whereas conventional vesicular stomatitis virus G (VSV-G)-LVs reached 5% at the most with H/F-LV entry occurring exclusively through the CD46 complement receptor. Importantly, reconstitution of NOD/SCIDγc-/- (NSG) mice with H/F-LV transduced prestimulated or resting hCD34+ cells confirmed these high transduction levels in all myeloid and lymphoid lineages. Remarkably, for resting CD34+ cells, secondary recipients exhibited increasing transduction levels of up to 100%, emphasizing that H/F-LVs efficiently gene-marked HSCs in the resting state. Because H/F-LVs promoted ex vivo gene modification of minimally manipulated CD34+ progenitors that maintained stemness, we assessed their applicability in Fanconi anemia, a bone marrow (BM) failure with chromosomal fragility. Notably, only H/F-LVs efficiently gene-corrected minimally stimulated hCD34+ cells in unfractionated BM from these patients. These H/F-LVs improved HSC gene delivery in the absence of cytokine stimulation while maintaining their stem cell potential. Thus, H/F-LVs will facilitate future clinical applications requiring HSC gene modification, including BM failure syndromes, for which treatment has been very challenging up to now.

10.
Mol Ther ; 23(11): 1734-1747, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26281898

ABSTRACT

The development of lentiviral vectors (LVs) for expression of a specific antibody can be achieved through the transduction of mature B-cells. This approach would provide a versatile tool for active immunotherapy strategies for infectious diseases or cancer, as well as for protein engineering. Here, we created a lentiviral expression system mimicking the natural production of these two distinct immunoglobulin isoforms. We designed a LV (FAM2-LV) expressing an anti-HCV-E2 surface glycoprotein antibody (AR3A) as a membrane-anchored Ig form or a soluble Ig form, depending on the B-cell maturation status. FAM2-LV induced high-level and functional membrane expression of the transgenic antibody in a nonsecretory B-cell line. In contrast, a plasma cell (PC) line transduced with FAM2-LV preferentially produced the secreted transgenic antibody. Similar results were obtained with primary B-cells transduced ex vivo. Most importantly, FAM2-LV transduced primary B-cells efficiently differentiated into PCs, which secreted the neutralizing anti-HCV E2 antibody upon adoptive transfer into immunodeficient NSG (NOD/SCIDγc(-/-)) recipient mice. Altogether, these results demonstrate that the conditional FAM2-LV allows preferential expression of the membrane-anchored form of an antiviral neutralizing antibody in B-cells and permits secretion of a soluble antibody following B-cell maturation into PCs in vivo.


Subject(s)
Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , Genetic Vectors , Immunoglobulin G/immunology , Lymphocyte Activation , Animals , Antibody-Dependent Cell Cytotoxicity , B-Lymphocytes/metabolism , Cell Line, Tumor , HEK293 Cells , Hepacivirus/immunology , Humans , Lentivirus , Membrane Glycoproteins/immunology , Mice , Mice, Inbred NOD , Protein Transport , Receptors, IgG/metabolism , Transduction, Genetic , Viral Envelope Proteins/immunology
11.
Blood ; 124(8): 1221-31, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-24951430

ABSTRACT

Hematopoietic stem cell (HSC)-based gene therapy holds promise for the cure of many diseases. The field is now moving toward the use of lentiviral vectors (LVs) as evidenced by 4 successful clinical trials. These trials used vesicular-stomatitis-virus-G protein (VSV-G)-LVs at high doses combined with strong cytokine-cocktail stimulation to obtain therapeutically relevant transduction levels; however, they might compromise the HSC character. Summarizing all these disadvantages, alternatives to VSV-G-LVs are urgently needed. We generated here high-titer LVs pseudotyped with a baboon retroviral envelope glycoprotein (BaEV-LVs), resistant to human complement. Under mild cytokine prestimulation to preserve the HSC characteristics, a single BaEV-LV application at a low dose, resulted in up to 90% of hCD34(+) cell transduction. Even more striking was that these new BaEV-LVs allowed, at low doses, efficient transduction of up to 30% of quiescent hCD34(+) cells, whereas high-dose VSV-G-LVs were insufficient. Importantly, reconstitution of NOD/Lt-SCID/γc(-/-) (NSG) mice with BaEV-LV-transduced hCD34(+) cells maintained these high transduction levels in all myeloid and lymphoid lineages, including early progenitors. This transduction pattern was confirmed or even increased in secondary NSG recipient mice. This suggests that BaEV-LVs efficiently transduce true HSCs and could improve HSC-based gene therapy, for which high-level HSC correction is needed for life-long cure.


Subject(s)
Betaretrovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Hematopoietic Stem Cells , Lentivirus/genetics , Transduction, Genetic , Viral Envelope Proteins/genetics , Animals , Antigens, CD34 , Cell Line , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Macaca , Membrane Glycoproteins/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
13.
Mol Ther ; 20(9): 1699-712, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22617109

ABSTRACT

Gene transfer into quiescent T and B cells is important for gene therapy and immunotherapy approaches. Previously, we generated lentiviral vectors (LVs) pseudotyped with Edmonston (Ed) measles virus (MV) hemagglutinin (H) and fusion (F) glycoproteins (H/F-LVs), which allowed efficient transduction of quiescent human T and B cells. However, a major obstacle in the use of H/F-LVs in vivo is that most of the human population is vaccinated against measles. As the MV humoral immune response is exclusively directed against the H protein of MV, we mutated the two dominant epitopes in H, Noose, and NE. LVs pseudotyped with these mutant H-glycoproteins escaped inactivation by monoclonal antibodies (mAbs) but were still neutralized by human serum. Consequently, we took advantage of newly emerged MV-D genotypes that were less sensitive to MV vaccination due to a different glycosylation pattern. The mutation responsible was introduced into the H/F-LVs, already mutated for Noose and NE epitopes. We found that these mutant H/F-LVs could efficiently transduce quiescent lymphocytes in the presence of high concentrations of MV antibody-positive human serum. Finally, upon incubation with total blood, mimicking the in vivo situation, the mutant H/F-LVs escaped MV antibody neutralization, where the original H/F-LVs failed. Thus, these novel H/F-LVs offer perspectives for in vivo lymphocyte-based gene therapy and immunotherapy.


Subject(s)
B-Lymphocytes/immunology , Lentivirus/genetics , Measles virus/genetics , T-Lymphocytes/immunology , Viral Fusion Proteins/genetics , Animals , Antibodies, Neutralizing , Antibodies, Viral , B-Lymphocytes/cytology , B-Lymphocytes/virology , Cell Line, Tumor , Cricetinae , Epitopes/genetics , Epitopes/immunology , Genetic Therapy , Genetic Vectors , Glycoproteins/genetics , Glycoproteins/immunology , Glycosylation , Hemagglutinins/genetics , Hemagglutinins/immunology , Humans , Immunity, Humoral , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/therapy , Immunotherapy , Lentivirus/immunology , Measles Vaccine/immunology , Measles virus/immunology , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/cytology , T-Lymphocytes/virology , Transduction, Genetic , Viral Fusion Proteins/immunology
14.
Blood ; 119(5): 1139-50, 2012 Feb 02.
Article in English | MEDLINE | ID: mdl-22117040

ABSTRACT

In vivo lentiviral vector (LV)-mediated gene delivery would represent a great step forward in the field of gene therapy. Therefore, we have engineered a novel LV displaying SCF and a mutant cat endogenous retroviral glycoprotein, RDTR. These RDTR/SCF-LVs outperformed RDTR-LVs for transduction of human CD34(+) cells (hCD34(+)). For in vivo gene therapy, these novel RDTR/SCF-displaying LVs can distinguish between the target hCD34(+) cells of interest and nontarget cells. Indeed, they selectively targeted transduction to 30%-40% of the hCD34(+) cells in cord blood mononuclear cells and in the unfractionated BM of healthy and Fanconi anemia donors, resulting in the correction of CD34(+) cells in the patients. Moreover, RDTR/SCF-LVs targeted transduction to CD34(+) cells with 95-fold selectivity compared with T cells in total cord blood. Remarkably, in vivo injection of the RDTR/SCF-LVs into the BM cavity of humanized mice resulted in the highly selective transduction of candidate hCD34(+)Lin(-) HSCs. In conclusion, this new LV will facilitate HSC-based gene therapy by directly targeting these primitive cells in BM aspirates or total cord blood. Most importantly, in the future, RDTR/SCF-LVs might completely obviate ex vivo handling and simplify gene therapy for many hematopoietic defects because of their applicability to direct in vivo inoculation.


Subject(s)
Bone Marrow/metabolism , Genetic Therapy/methods , Genetic Vectors/physiology , Hematopoietic Stem Cells/metabolism , Hemoglobinuria, Paroxysmal/therapy , Lentivirus/genetics , Anemia, Aplastic , Animals , Animals, Newborn , Bone Marrow/pathology , Bone Marrow Diseases , Bone Marrow Failure Disorders , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Targeting/methods , Gene Transfer Techniques , Genetic Vectors/genetics , Hemoglobinuria, Paroxysmal/genetics , Hemoglobinuria, Paroxysmal/pathology , Humans , Immunoglobulin gamma-Chains/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Transduction, Genetic
15.
J Virol ; 85(12): 5975-85, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21450813

ABSTRACT

Gene transfer into quiescent T and B cells is of importance for gene therapy and immunotherapy approaches to correct hematopoietic disorders. Previously, we generated lentiviral vectors (LVs) pseudotyped with the Edmonston measles virus (MV) hemagglutinin and fusion glycoproteins (Hgps and Fgps) (H/F-LVs), which, for the first time, allowed efficient transduction of quiescent human B and T cells. These target cells express both MV entry receptors used by the vaccinal Edmonston strain, CD46 and signaling lymphocyte activation molecule (SLAM). Interestingly, LVs pseudotyped with an MV Hgp, blind for the CD46 binding site, were completely inefficient for resting-lymphocyte transduction. Similarly, SLAM-blind H mutants that recognize only CD46 as the entry receptor did not allow stable LV transduction of resting T cells. The CD46-tropic LVs accomplished vector-cell binding, fusion, entry, and reverse transcription at levels similar to those achieved by the H/F-LVs, but efficient proviral integration did not occur. Our results indicate that both CD46 and SLAM binding sites need to be present in cis in the Hgp to allow successful stable transduction of quiescent lymphocytes. Moreover, the entry mechanism utilized appears to be crucial: efficient transduction was observed only when CD46 and SLAM were correctly engaged and an entry mechanism that strongly resembles macropinocytosis was triggered. Taken together, our results suggest that although vector entry can occur through the CD46 receptor, SLAM binding and subsequent signaling are also required for efficient LV transduction of quiescent lymphocytes to occur.


Subject(s)
Antigens, CD/metabolism , Gene Transfer Techniques , Genetic Vectors/genetics , Glycoproteins/genetics , Lentivirus/genetics , Lymphocyte Activation , Measles virus/genetics , Membrane Cofactor Protein/metabolism , Receptors, Cell Surface/metabolism , Adult , Antigens, CD/genetics , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Humans , Lentivirus/metabolism , Measles virus/chemistry , Membrane Cofactor Protein/genetics , Pinocytosis , Receptors, Cell Surface/genetics , Signaling Lymphocytic Activation Molecule Family Member 1 , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Transduction, Genetic , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...