Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
Cell Rep ; 38(3): 110279, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35045303

ABSTRACT

Murine leukemia virus (MLV)-presenting cells form stable intercellular contacts with target cells during infection of lymphoid tissue, indicating a role of cell-cell contacts in retrovirus dissemination. Whether host cell adhesion proteins are required for retrovirus spread in vivo remains unknown. Here, we demonstrate that the lymphocyte-function-associated-antigen-1 (LFA1) and its ligand intercellular-adhesion-molecule-1 (ICAM1) are important for cell-contact-dependent transmission of MLV between leukocytes. Infection experiments in LFA1- and ICAM1-deficient mice demonstrate a defect in MLV spread within lymph nodes. Co-culture of primary leukocytes reveals a specific requirement for ICAM1 on donor cells and LFA1 on target cells for cell-contact-dependent spread through trans- and cis-infection. Importantly, adoptive transfer experiments combined with a newly established MLV-fusion assay confirm that the directed LFA1-ICAM1 interaction is important for retrovirus fusion and transmission in vivo. Taken together, our data provide insights on how retroviruses exploit host proteins and the biology of cell-cell interactions for dissemination.


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/virology , Lymphocyte Function-Associated Antigen-1/metabolism , Retroviridae Infections/virology , Animals , Host-Pathogen Interactions/physiology , Lymphocytes/virology , Macrophages/virology , Mice , Mice, Inbred C57BL , Retroviridae Infections/transmission , Tumor Virus Infections/transmission , Tumor Virus Infections/virology
2.
J Mol Biol ; 433(18): 167111, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34153286

ABSTRACT

5-aza-cytidine (5-aza-C) has been shown to be a potent human immunodeficiency virus type 1 (HIV-1) mutagen that induces G-to-C hypermutagenesis by incorporation of the reduced form (i.e., 5-aza-dC, 5-aza-dCTP). Evidence to date suggests that this lethal mutagenesis is the primary antiretroviral mechanism for 5-aza-C. To investigate the breadth of application of 5-aza-C as an antiretroviral mutagen, we have conducted a comparative, parallel analysis of the antiviral mechanism of 5-aza-C between HIV-1 and gammaretroviruses - i.e., murine leukemia virus (MuLV) and feline leukemia virus (FeLV). Intriguingly, in contrast to the hallmark G-to-C hypermutagenesis observed with HIV-1, MuLV and FeLV did not reveal the presence of a significant increase in mutational burden, particularly that of G-to-C transversion mutations. The effect of 5-aza-dCTP on DNA synthesis revealed that while HIV-1 RT was not inhibited by 5-aza-dCTP even at 100 µM, 5-aza-dCTP was incorporated and significantly inhibited MuLV RT, generating pause sites and reducing the fully extended product. 5-aza-dCTP was found to be incorporated into DNA by MuLV RT or HIV-1 RT, but only acted as a non-obligate chain terminator for MuLV RT. This biochemical data provides an independent line of experimental evidence in support of the conclusion that HIV-1 and MuLV have distinct primary mechanisms of antiretroviral action with 5-aza-C. Taken together, our data provides striking evidence that an antiretroviral mutagen can have strong potency via distinct mechanisms of action among closely related viruses, unlinking antiviral activity from antiviral mechanism of action.


Subject(s)
Antiviral Agents/pharmacology , Azacitidine/analogs & derivatives , Cytidine Triphosphate/analogs & derivatives , HIV Infections/drug therapy , Leukemia, Experimental/drug therapy , Mutation/drug effects , Retroviridae Infections/drug therapy , Tumor Virus Infections/drug therapy , Animals , Azacitidine/pharmacology , Cats , Cytidine Triphosphate/pharmacology , HIV/drug effects , HIV Infections/virology , Humans , Leukemia Virus, Feline/drug effects , Leukemia Virus, Murine/drug effects , Leukemia, Experimental/virology , Mice , Mutagenesis , Mutagens , Retroviridae Infections/virology , Tumor Virus Infections/virology , Virus Replication
3.
J Ethnopharmacol ; 267: 113519, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33137432

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Astragalus glycyphyllos L. has been extensively used in Bulgarian folk medicine as an antihypertensive, diuretic, anti-inflammatory, anti-tumour, in cases of cardiac insufficiency, renal inflammation, calculosis, etc. AIM OF THE STUDY: To evaluate the possible in vitro/in vivo anti-proliferative/anti-tumour activity of a purified saponins' mixture (PSM) obtained from the plant. MATERIALS AND METHODS: Viability and proliferative activity of the Graffi myeloid tumour cells was assessed by MTT test. The morphological alterations were visualized and analysed by fluorescent microscopy after intravital double staining. An in vivo model of Graffi tumour bearing hamsters was used to examine the influence of PSM on transplantability, tumour growth, survival and mortality as well as to observe pathomorphological changes. RESULTS: Graffi tumour cells were sensitive to purified saponins' mixture after 24 and 48 h treatment. The treatment induced a statistically significant decrease of the viability/proliferation of the Graffi tumour cells. These effects were concentration- and time-dependent. Fluorescent microscopy studies showed that these antiproliferative effects were connected to the induction of apoptosis. The in vivo study showed the presence of a stromal component, single mononuclear cells in the stroma. Multiple incorrect mitotic figures were observed in the tumour tissue from the control group. Well-formed stroma with accumulation of mononuclear cells and mitotic cells were found in the group, treated with PSM. The tumour weight was decreased in the group, treated with PMS. CONCLUSION: The results indicate that PSM exhibited in vitro/in vivo antiproliferative/anti-tumour effects.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Astragalus Plant , Cell Proliferation/drug effects , Leukemia, Experimental/drug therapy , Plant Extracts/pharmacology , Saponins/pharmacology , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Astragalus Plant/chemistry , Cricetinae , Female , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/pathology , Leukemia, Experimental/virology , Male , Mesocricetus , Plant Extracts/isolation & purification , Primary Cell Culture , Retroviridae Infections/virology , Saponins/isolation & purification , Tumor Burden/drug effects , Tumor Cells, Cultured , Tumor Virus Infections/virology
4.
Blood ; 137(7): 908-922, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33174606

ABSTRACT

Epigenetic regulation is essential for the maintenance of the hematopoietic system, and its deregulation is implicated in hematopoietic disorders. In this study, UTX, a demethylase for lysine 27 on histone H3 (H3K27) and a component of COMPASS-like and SWI/SNF complexes, played an essential role in the hematopoietic system by globally regulating aging-associated genes. Utx-deficient (UtxΔ/Δ) mice exhibited myeloid skewing with dysplasia, extramedullary hematopoiesis, impaired hematopoietic reconstituting ability, and increased susceptibility to leukemia, which are the hallmarks of hematopoietic aging. RNA-sequencing (RNA-seq) analysis revealed that Utx deficiency converted the gene expression profiles of young hematopoietic stem-progenitor cells (HSPCs) to those of aged HSPCs. Utx expression in hematopoietic stem cells declined with age, and UtxΔ/Δ HSPCs exhibited increased expression of an aging-associated marker, accumulation of reactive oxygen species, and impaired repair of DNA double-strand breaks. Pathway and chromatin immunoprecipitation analyses coupled with RNA-seq data indicated that UTX contributed to hematopoietic homeostasis mainly by maintaining the expression of genes downregulated with aging via demethylase-dependent and -independent epigenetic programming. Of note, comparison of pathway changes in UtxΔ/Δ HSPCs, aged muscle stem cells, aged fibroblasts, and aged induced neurons showed substantial overlap, strongly suggesting common aging mechanisms among different tissue stem cells.


Subject(s)
Aging/genetics , Gene Expression Regulation/genetics , Hematopoiesis/genetics , Hematopoietic System/physiology , Histone Code/genetics , Histone Demethylases/physiology , Animals , Cellular Senescence/genetics , DNA Breaks, Double-Stranded , DNA Repair , Female , Genetic Predisposition to Disease , Hematopoiesis, Extramedullary , Histone Demethylases/deficiency , Histone Demethylases/genetics , Immune Reconstitution , Jumonji Domain-Containing Histone Demethylases/metabolism , Leukemia, Experimental/genetics , Leukemia, Experimental/virology , Male , Mice , Mice, Knockout , Moloney murine leukemia virus/physiology , Myeloid Cells/pathology , Radiation Chimera , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism , Transcription Factors/metabolism , Virus Integration
5.
Biochim Biophys Acta Mol Basis Dis ; 1866(12): 165922, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32800945

ABSTRACT

Excessive production of immunoglobulins (Ig) causes endoplasmic reticulum (ER) stress and triggers the unfolded protein response (UPR). Hypergammaglobulinemia and lymphadenopathy are hallmarks of murine AIDS that develops in mice infected with the LP-BM5 murine leukemia retrovirus complex. In these mice, Th2 polarization and aberrant humoral response have been previously correlated to altered intracellular redox homeostasis. Our goal was to understand the role of the cell's redox state in Ig secretion and plasma cell (PC) maturation. To this aim, LP-BM5-infected mice were treated with I-152, an N-acetyl-cysteine and cysteamine supplier. Intraperitoneal I-152 administration (30 µmol/mouse three times a week for 9 weeks) decreased plasma IgG and increased IgG/Syndecan 1 ratio in the lymph nodes where IgG were in part accumulated within the ER. PC containing cytoplasmic inclusions filled with IgG were present in all animals, with fewer mature PC in those treated with I-152. Infection induced up-regulation of signaling molecules involved in the UPR, i.e. CHAC1, BiP, sXBP-1 and PDI, that were generally unaffected by I-152 treatment except for PDI and sXBP-1, which have a key role in protein folding and PC maturation, respectively. Our data suggest that one of the mechanisms through which I-152 can limit hypergammaglobulinemia in LP-BM5-infected mice is by influencing IgG folding/assembly as well as secretion and affecting PC maturation.


Subject(s)
Acetylcysteine/analogs & derivatives , Antiviral Agents/pharmacology , Cysteamine/analogs & derivatives , Immunoglobulins/metabolism , Plasma Cells/drug effects , Retroviridae Infections/drug therapy , Tumor Virus Infections/drug therapy , Unfolded Protein Response/drug effects , Acetylcysteine/administration & dosage , Acetylcysteine/pharmacology , Animals , Antiviral Agents/administration & dosage , Cysteamine/administration & dosage , Cysteamine/pharmacology , Disease Models, Animal , Female , Immunoglobulins/blood , Injections, Intraperitoneal , Leukemia, Experimental/drug therapy , Leukemia, Experimental/metabolism , Leukemia, Experimental/virology , Mice , Mice, Inbred C57BL , Plasma Cells/metabolism , Plasma Cells/virology , Protein Unfolding/drug effects , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology
6.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32641479

ABSTRACT

Apolipoprotein B editing enzyme, catalytic polypeptide 3 (APOBEC3) family members are cytidine deaminases that play important roles in intrinsic responses to retrovirus infection. Complex retroviruses like human immunodeficiency virus type 1 (HIV-1) encode the viral infectivity factor (Vif) protein to counteract APOBEC3 proteins. Vif induces degradation of APOBEC3G and other APOBEC3 proteins and thereby prevents their packaging into virions. It is not known if murine leukemia virus (MLV) encodes a Vif-like protein. Here, we show that the MLV P50 protein, produced from an alternatively spliced gag RNA, interacts with the C terminus of mouse APOBEC3 and prevents its packaging without causing its degradation. By infecting APOBEC3 knockout (KO) and wild-type (WT) mice with Friend or Moloney MLV P50-deficient viruses, we found that APOBEC3 restricts the mutant viruses more than WT viruses in vivo Replication of P50-mutant viruses in an APOBEC3-expressing stable cell line was also much slower than that of WT viruses, and overexpressing P50 in this cell line enhanced mutant virus replication. Thus, MLV encodes a protein, P50, that overcomes APOBEC3 restriction by preventing its packaging into virions.IMPORTANCE MLV has existed in mice for at least a million years, in spite of the existence of host restriction factors that block infection. Although MLV is considered a simple retrovirus compared to lentiviruses, it does encode proteins generated from alternatively spliced RNAs. Here, we show that P50, generated from an alternatively spliced RNA encoded in gag, counteracts APOBEC3 by blocking its packaging. MLV also encodes a protein, glycoGag, that increases capsid stability and limits APOBEC3 access to the reverse transcription complex (RTC). Thus, MLV has evolved multiple means of preventing APOBEC3 from blocking infection, explaining its survival as an infectious pathogen in mice.


Subject(s)
Cytidine Deaminase/genetics , Gene Expression Regulation, Viral , Gene Products, gag/genetics , Leukemia, Experimental/genetics , Moloney murine leukemia virus/genetics , Retroviridae Infections/genetics , Tumor Virus Infections/genetics , Alternative Splicing , Animals , Capsid/metabolism , Cytidine Deaminase/deficiency , Gene Products, gag/metabolism , HEK293 Cells , Host-Pathogen Interactions/genetics , Humans , Leukemia, Experimental/metabolism , Leukemia, Experimental/virology , Mice , Mice, Knockout , Moloney murine leukemia virus/metabolism , Moloney murine leukemia virus/pathogenicity , NIH 3T3 Cells , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Signal Transduction , Tumor Virus Infections/metabolism , Tumor Virus Infections/virology , Virion/genetics , Virion/metabolism , Virion/pathogenicity , Virus Replication
7.
mBio ; 11(4)2020 07 14.
Article in English | MEDLINE | ID: mdl-32665269

ABSTRACT

The serine incorporator (SERINC) proteins are multipass transmembrane proteins that affect sphingolipid and phosphatidylserine synthesis. Human SERINC5 and SERINC3 were recently shown to possess antiretroviral activity for a number of retroviruses, including human immunodeficiency virus (HIV), murine leukemia virus (MLV), and equine infectious anemia virus (EIAV). In the case of MLV, the glycosylated Gag (glyco-Gag) protein was shown to counteract SERINC5-mediated restriction in in vitro experiments and the viral envelope was found to determine virion sensitivity or resistance to SERINC5. However, nothing is known about the in vivo function of SERINC5. Antiretroviral function of a host factor in vitro is not always associated with antiretroviral function in vivo Using SERINC5-/- mice that we had generated, we showed that mouse SERINC5 (mSERINC5) restriction of MLV infection in vivo is influenced not only by glyco-Gag but also by the retroviral envelope. Finally, we also examined the in vivo function of the other SERINC gene with known antiretroviral functions, SERINC3. By using SERINC3-/- mice, we found that the murine homologue, mSERINC3, had no antiretroviral role either in vivo or in vitro To our knowledge, this report provides the first data showing that SERINC5 restricts retrovirus infection in vivo and that restriction of retrovirus infectivity in vivo is dependent on the presence of both glyco-Gag and the viral envelope.IMPORTANCE This study examined for the first time the in vivo function of the serine incorporator (SERINC) proteins during retrovirus infection. SERINC3 and SERINC5 (SERINC3/5) restrict a number of retroviruses, including human immunodeficiency virus 1 (HIV-1) and murine leukemia virus (MLV), by blocking their entry into cells. Nevertheless, HIV-1 and MLV encode factors, Nef and glycosylated Gag, respectively, that counteract SERINC3/5 in vitro We recently developed SERINC3 and SERINC5 knockout mice to examine the in vivo function of these genes. We found that SERINC5 restriction is dependent on the absence of glycosylated Gag and the expression of a specific viral envelope glycoprotein. On the other hand, SERINC3 had no antiviral function. Our findings have implications for the development of therapeutics that target SERINC5 during retrovirus infection.


Subject(s)
Host-Pathogen Interactions , Leukemia, Experimental/virology , Membrane Proteins/genetics , Retroviridae Infections/virology , Tumor Virus Infections/virology , Animals , Female , Glycosylation , Leukemia Virus, Murine/pathogenicity , Male , Membrane Glycoproteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
mBio ; 10(1)2019 02 19.
Article in English | MEDLINE | ID: mdl-30782653

ABSTRACT

B cell follicles of the spleen and lymph nodes are immune privileged sites and serve as sanctuaries for infected CD4+ cells in HIV infection. It is assumed that CD8+ T cell responses promote the establishment of the reservoir, as B cell follicles do not permit CD8+ T cell entry. Here we analyzed the infected cell population in the Friend retrovirus (FV) infection and investigated whether FV can similarly infect follicular cells. For analysis of FV-infected cells, we constructed a recombinant FV encoding the bright fluorescent protein mWasabi and performed flow cytometry with cells isolated from spleens, lymph nodes and bone marrow of FV-mWasabi-infected mice. Using t-stochastic neighbor embedding for data exploration, we demonstrate how the target cell population changes during the course of infection. While FV was widely distributed in erythrocytes, myeloid cells, B cells, and CD4+ T cells in the acute phase of infection, the bulk viral load in the late phase was carried by macrophages and follicular B and CD4+ T cells, suggesting that FV persists in cells that are protected from CD8+ T cell killing. Importantly, seeding into follicular cells was equally observed in CD8+ T cell-depleted mice and in highly FV-susceptible mice that mount a very weak immune response, demonstrating that infection of follicular cells is not driven by immune pressure. Our data demonstrate that infection of cells in the B cell follicle is a characteristic of the FV infection, making this murine retrovirus an even more valuable model for development of retrovirus immunotherapy approaches.IMPORTANCE Human immunodeficiency virus is notorious for its ability to avoid clearance by therapeutic interventions, which is partly attributed to the establishment of reservoirs in latently infected cells and cells that reside in immunologically privileged B cell follicles. In the work presented here, we show that cells of the B cell follicle are equally infected by a simple mouse gammaretrovirus. Using fluorescently labeled Friend retrovirus, we found that B cells and T cells in the B cell follicle, while not carrying the bulk of the virus load, were indeed infected by Friend virus in the early acute phase of the infection and persisted in the chronic infection. Our results suggest that infection of follicular cells may be a shared property of lymphotropic viruses and propose the FV infection of mice as a useful model to study strategies for follicular reservoir elimination.


Subject(s)
B-Lymphocytes/virology , Friend murine leukemia virus/growth & development , Leukemia, Experimental/virology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Animals , Flow Cytometry , Friend murine leukemia virus/genetics , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Lymph Nodes/virology , Mice , Spleen/virology , Staining and Labeling
9.
mBio ; 10(1)2019 01 22.
Article in English | MEDLINE | ID: mdl-30670616

ABSTRACT

Friend virus (FV) is a naturally occurring mouse retrovirus that infects dividing cells of the hematopoietic lineage, including antigen-presenting cells (APCs). The infection of APCs by viruses often induces their dysfunction, and it has been shown that FV infection reduces the ability of dendritic cells (DCs) to prime critical CD8+ T cell responses. Nonetheless, mice mount vigorous CD8+ T cell responses, so we investigated whether B cells might serve as alternative APCs during FV infection. Direct ex vivo analysis of B cells from FV-infected mice revealed that infected but not uninfected B cells upregulated expression of the costimulatory molecules CD80, CD86, and CD40, as well as major histocompatibility complex class II (MHC-II) molecules. Furthermore, in vitro studies showed that, compared to uninfected B cells from the same mice, the FV-infected B cells had significantly enhanced APC function, as measured by their capacity to prime CD8+ T cell activation and proliferation. Thus, in contrast to DCs, infection of B cells with FV enhanced their APC capacity and ability to stimulate the CD8+ T cell responses essential for virus control. FV infections also induce the activation and expansion of regulatory T cells (Tregs), so it was of interest to determine the impact of Tregs on B cell activation. The upregulation of costimulatory molecule expression and APC function of B cells was even more strongly enhanced by in vivo depletion of regulatory T cells than infection. Thus, Tregs exert potent homeostatic suppression of B cell activation that is partially overcome by FV infection.IMPORTANCE The primary role of B cells in immunity is considered the production of pathogen-specific antibodies, but another, less-well-studied, function of B cells is to present foreign antigens to T cells to stimulate their activation and proliferation. Dendritic cells (DCs) are considered the most important antigen-presenting cells (APCs) for CD8+ T cells, but DCs lose APC function when infected with Friend virus (FV), a model retrovirus of mice. Interestingly, B cells were better able to stimulate CD8+ T cell responses when they were infected with FV. We also found that the activation status of B cells under homeostatic conditions was potently modulated by regulatory T cells. This study illustrates an important link between B cell and T cell responses and illustrates an additional mechanism by which regulatory T cells suppress critical T cell responses during viral infections.


Subject(s)
Antigen Presentation , B-Lymphocytes/immunology , Friend murine leukemia virus/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes/chemistry , B7-1 Antigen/analysis , B7-2 Antigen/analysis , CD40 Antigens/analysis , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Histocompatibility Antigens Class II/analysis , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Lymphocyte Activation , Mice , Retroviridae Infections/immunology , Retroviridae Infections/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
10.
Exp Hematol ; 68: 2-9, 2018 12.
Article in English | MEDLINE | ID: mdl-30391350

ABSTRACT

The transcription factor RUNX1 is required in the embryo for formation of the adult hematopoietic system. Here, we describe the seminal findings that led to the discovery of RUNX1 and of its critical role in blood cell formation in the embryo from hemogenic endothelium (HE). We also present RNA-sequencing data demonstrating that HE cells in different anatomic sites, which produce hematopoietic progenitors with dissimilar differentiation potentials, are molecularly distinct. Hemogenic and non-HE cells in the yolk sac are more closely related to each other than either is to hemogenic or non-HE cells in the major arteries. Therefore, a major driver of the different lineage potentials of the committed erythro-myeloid progenitors that emerge in the yolk sac versus hematopoietic stem cells that originate in the major arteries is likely to be the distinct molecular properties of the HE cells from which they are derived. We used bioinformatics analyses to predict signaling pathways active in arterial HE, which include the functionally validated pathways Notch, Wnt, and Hedgehog. We also used a novel bioinformatics approach to assemble transcriptional regulatory networks and predict transcription factors that may be specifically involved in hematopoietic cell formation from arterial HE, which is the origin of the adult hematopoietic system.


Subject(s)
Core Binding Factor Alpha 2 Subunit/physiology , Hemangioblasts/physiology , Hematopoiesis/physiology , Animals , Arteries/cytology , Arteries/embryology , Core Binding Factor Alpha 2 Subunit/deficiency , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor beta Subunit/deficiency , Core Binding Factor beta Subunit/genetics , Core Binding Factor beta Subunit/physiology , Drosophila Proteins/genetics , Fetal Blood/physiology , Gene Expression Regulation, Developmental , Humans , Leukemia, Experimental/genetics , Leukemia, Experimental/virology , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Knockout , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Transcription, Genetic , Yolk Sac/cytology
11.
J Virol ; 91(21)2017 11 01.
Article in English | MEDLINE | ID: mdl-28794032

ABSTRACT

Ecotropic, xenotropic, and polytropic mouse leukemia viruses (E-, X-, and P-MLVs) exist in mice as infectious viruses and endogenous retroviruses (ERVs) inserted into mouse chromosomes. All three MLV subgroups are linked to leukemogenesis, which involves generation of recombinants with polytropic host range. Although P-MLVs are deemed to be the proximal agents of disease induction, few biologically characterized infectious P-MLVs have been sequenced for comparative analysis. We analyzed the complete genomes of 16 naturally occurring infectious P-MLVs, 12 of which were typed for pathogenic potential. We sought to identify ERV progenitors, recombinational hot spots, and segments that are always replaced, never replaced, or linked to pathogenesis or host range. Each P-MLV has an E-MLV backbone with P- or X-ERV replacements that together cover 100% of the recombinant genomes, with different substitution patterns for X- and P-ERVs. Two segments are always replaced, both coding for envelope (Env) protein segments: the N terminus of the surface subunit and the cytoplasmic tail R peptide. Viral gag gene replacements are influenced by host restriction genes Fv1 and Apobec3 Pathogenic potential maps to the env transmembrane subunit segment encoding the N-heptad repeat (HR1). Molecular dynamics simulations identified three novel interdomain salt bridges in the lymphomagenic virus HR1 that could affect structural stability, entry or sensitivity to host immune responses. The long terminal repeats of lymphomagenic P-MLVs are differentially altered by recombinations, duplications, or mutations. This analysis of the naturally occurring, sometimes pathogenic P-MLV recombinants defines the limits and extent of intersubgroup recombination and identifies specific sequence changes linked to pathogenesis and host interactions.IMPORTANCE During virus-induced leukemogenesis, ecotropic mouse leukemia viruses (MLVs) recombine with nonecotropic endogenous retroviruses (ERVs) to produce polytropic MLVs (P-MLVs). Analysis of 16 P-MLV genomes identified two segments consistently replaced: one at the envelope N terminus that alters receptor choice and one in the R peptide at the envelope C terminus, which is removed during virus assembly. Genome-wide analysis shows that nonecotropic replacements in the progenitor ecotropic MLV genome are more extensive than previously appreciated, covering 100% of the genome; contributions from xenotropic and polytropic ERVs differentially alter the regions responsible for receptor determination or subject to APOBEC3 and Fv1 restriction. All pathogenic viruses had modifications in the regulatory elements in their long terminal repeats and differed in a helical segment of envelope involved in entry and targeted by the host immune system. Virus-induced leukemogenesis thus involves generation of complex recombinants, and specific replacements are linked to pathogenesis and host restrictions.


Subject(s)
Host Specificity/genetics , Leukemia Virus, Murine/classification , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/virology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Viral Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , Evolution, Molecular , Genome, Viral , Leukemia Virus, Murine/genetics , Mice , Molecular Dynamics Simulation , Protein Conformation , Receptors, Virus/genetics , Receptors, Virus/metabolism , Sequence Homology , Terminal Repeat Sequences , Viral Proteins/chemistry , Viral Proteins/metabolism
12.
In Vivo ; 31(3): 313-320, 2017.
Article in English | MEDLINE | ID: mdl-28438857

ABSTRACT

BACKGROUND/AIM: The polycythemia form of Friend leukemia virus (FVP) causes splenomegaly and lethal erythroleukemia in Fv-2s-susceptible mouse strains. We sought to determine whether the hematopoietic stem cell (HSC) pool was expanded in Fv-2r-resistant mice. MATERIALS AND METHODS: The 120-day bone marrow transplantation competitive repopulation assay was used to determine whether FVP-infected Fv-2r C57BL/6 mice demonstrated expansion of the HSC pool compared to the pool of committed hematopoietic progenitor cells in the same marrow assayed in vitro. RESULTS: There was a significant expansion of committed hematopoietic progenitors observed in virus-infected Fv-2s FVB mice, but not Fv-2r C57BL/6 mice. Furthermore, Fv-2r mice showed no detectable expansion of either committed hematopoietic progenitor cells or the multipotential stem cell pool by competitive repopulation assay. CONCLUSION: Friend virus disease in Fv-2s mice is associated with expansion of committed hematopoietic progenitors. Fv-2r mice show no expansion of either committed progenitor or pluripotential stem cell numbers.


Subject(s)
Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Retroviridae Infections/pathology , Tumor Virus Infections/pathology , Animals , Bone Marrow/virology , Female , Friend murine leukemia virus/pathogenicity , Hematopoietic Stem Cells/virology , Leukemia, Erythroblastic, Acute/pathology , Leukemia, Erythroblastic, Acute/virology , Leukemia, Experimental/pathology , Leukemia, Experimental/virology , Male , Mice , Mice, Inbred C57BL , Retroviridae Infections/virology , Spleen/pathology , Spleen/virology , Splenomegaly/pathology , Splenomegaly/virology , Tumor Virus Infections/virology
13.
J Virol ; 90(16): 7118-7130, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27226373

ABSTRACT

UNLABELLED: Injection of the LP-BM5 murine leukemia virus into mice causes murine AIDS, a disease characterized by many dysfunctions of immunocompetent cells. To establish whether the disease is characterized by glutathione imbalance, reduced glutathione (GSH) and cysteine were quantified in different organs. A marked redox imbalance, consisting of GSH and/or cysteine depletion, was found in the lymphoid organs, such as the spleen and lymph nodes. Moreover, a significant decrease in cysteine and GSH levels in the pancreas and brain, respectively, was measured at 5 weeks postinfection. The Th2 immune response was predominant at all times investigated, as revealed by the expression of Th1/Th2 cytokines. Furthermore, investigation of the activation status of peritoneal macrophages showed that the expression of genetic markers of alternative activation, namely, Fizz1, Ym1, and Arginase1, was induced. Conversely, expression of inducible nitric oxide synthase, a marker of classical activation of macrophages, was detected only when Th1 cytokines were expressed at high levels. In vitro studies revealed that during the very early phases of infection, GSH depletion and the downregulation of interleukin-12 (IL-12) p40 mRNA were correlated with the dose of LP-BM5 used to infect the macrophages. Treatment of LP-BM5-infected mice with N-(N-acetyl-l-cysteinyl)-S-acetylcysteamine (I-152), an N-acetyl-cysteine supplier, restored GSH/cysteine levels in the organs, reduced the expression of alternatively activated macrophage markers, and increased the level of gamma interferon production, while it decreased the levels of Th2 cytokines, such as IL-4 and IL-5. Our findings thus establish a link between GSH deficiency and Th1/Th2 disequilibrium in LP-BM5 infection and indicate that I-152 can be used to restore the GSH level and a balanced Th1/Th2 response in infected mice. IMPORTANCE: The first report of an association between Th2 polarization and alteration of the redox state in LP-BM5 infection is presented. Moreover, it provides evidence that LP-BM5 infection causes a decrease in the thiol content of peritoneal macrophages, which can influence IL-12 production. The restoration of GSH levels by GSH-replenishing molecules can represent a new therapeutic avenue to fight this retroviral infection, as it reestablishes the Th1/Th2 balance. Immunotherapy based on the use of pro-GSH molecules would permit LP-BM5 infection and probably all those viral infections characterized by GSH deficiency and a Th1/Th2 imbalance to be more effectively combated.


Subject(s)
Glutathione/deficiency , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/complications , Murine Acquired Immunodeficiency Syndrome/etiology , Retroviridae Infections/complications , Th2 Cells/immunology , Tumor Virus Infections/complications , Animals , Cells, Cultured , Cytokines/metabolism , Female , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Lymphocyte Activation , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/virology , Mice , Mice, Inbred C57BL , Murine Acquired Immunodeficiency Syndrome/metabolism , Murine Acquired Immunodeficiency Syndrome/pathology , Retroviridae Infections/immunology , Retroviridae Infections/virology , Spleen/immunology , Spleen/metabolism , Spleen/virology , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/virology , Th2 Cells/metabolism , Th2 Cells/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/virology
14.
J Virol ; 90(15): 6896-6905, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27194765

ABSTRACT

UNLABELLED: During the early steps of infection, retroviruses must direct the movement of the viral genome into the nucleus to complete their replication cycle. This process is mediated by cellular proteins that interact first with the reverse transcription complex and later with the preintegration complex (PIC), allowing it to reach and enter the nucleus. For simple retroviruses, such as murine leukemia virus (MLV), the identities of the cellular proteins involved in trafficking of the PIC in infection are unknown. To identify cellular proteins that interact with the MLV PIC, we developed a replication-competent MLV in which the integrase protein was tagged with a FLAG epitope. Using a combination of immunoprecipitation and mass spectrometry, we established that the microtubule motor dynein regulator DCTN2/p50/dynamitin interacts with the MLV preintegration complex early in infection, suggesting a direct interaction between the incoming viral particles and the dynein complex regulators. Further experiments showed that RNA interference (RNAi)-mediated silencing of either DCTN2/p50/dynamitin or another dynein regulator, NudEL, profoundly reduced the efficiency of infection by ecotropic, but not amphotropic, MLV reporters. We propose that the cytoplasmic dynein regulators are a critical component of the host machinery needed for infection by the retroviruses entering the cell via the ecotropic envelope pathway. IMPORTANCE: Retroviruses must access the chromatin of host cells to integrate the viral DNA, but before this crucial event, they must reach the nucleus. The movement through the cytoplasm-a crowded environment where diffusion is slow-is thought to utilize retrograde transport along the microtubule network by the dynein complex. Different viruses use different components of this multisubunit complex. We found that the preintegration complex of murine leukemia virus (MLV) interacts with the dynein complex and that regulators of this complex are essential for infection. Our study provides the first insight into the requirements for retrograde transport of the MLV preintegration complex.


Subject(s)
Dyneins/metabolism , Leukemia Virus, Murine/physiology , Leukemia, Experimental/virology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Animals , Genome, Viral , Leukemia, Experimental/metabolism , Mice , NIH 3T3 Cells , Retroviridae Infections/metabolism , Tumor Virus Infections/metabolism
15.
J Biol Chem ; 290(45): 27345-27359, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26370074

ABSTRACT

Siglec-1 (sialoadhesin, CD169) is a surface receptor on human cells that mediates trans-enhancement of HIV-1 infection through recognition of sialic acid moieties in virus membrane gangliosides. Here, we demonstrate that mouse Siglec-1, expressed on the surface of primary macrophages in an interferon-α-responsive manner, captures murine leukemia virus (MLV) particles and mediates their transfer to proliferating lymphocytes. The MLV infection of primary B-cells was markedly more efficient than that of primary T-cells. The major structural protein of MLV particles, Gag, frequently co-localized with Siglec-1, and trans-infection, primarily of surface-bound MLV particles, efficiently occurred. To explore the role of sialic acid for MLV trans-infection at a submolecular level, we analyzed the potential of six sialic acid precursor analogs to modulate the sialylated ganglioside-dependent interaction of MLV particles with Siglec-1. Biosynthetically engineered sialic acids were detected in both the glycolipid and glycoprotein fractions of MLV producer cells. MLV released from cells carrying N-acyl-modified sialic acids displayed strikingly different capacities for Siglec-1-mediated capture and trans-infection; N-butanoyl, N-isobutanoyl, N-glycolyl, or N-pentanoyl side chain modifications resulted in up to 92 and 80% reduction of virus particle capture and trans-infection, respectively, whereas N-propanoyl or N-cyclopropylcarbamyl side chains had no effect. In agreement with these functional analyses, molecular modeling indicated reduced binding affinities for non-functional N-acyl modifications. Thus, Siglec-1 is a key receptor for macrophage/lymphocyte trans-infection of surface-bound virions, and the N-acyl side chain of sialic acid is a critical determinant for the Siglec-1/MLV interaction.


Subject(s)
Moloney murine leukemia virus/pathogenicity , Sialic Acid Binding Ig-like Lectin 1/chemistry , Sialic Acid Binding Ig-like Lectin 1/physiology , Animals , Binding Sites , Cell Line , Gangliosides/chemistry , Gangliosides/metabolism , Host-Pathogen Interactions/physiology , Humans , Interferon-alpha/physiology , Leukemia, Experimental/physiopathology , Leukemia, Experimental/virology , Lymphocytes/physiology , Lymphocytes/virology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/physiology , N-Acetylneuraminic Acid/chemistry , Receptors, Virus/chemistry , Receptors, Virus/physiology , Retroviridae Infections/physiopathology , Retroviridae Infections/virology , Sialic Acid Binding Ig-like Lectin 1/genetics , Tumor Virus Infections/physiopathology , Tumor Virus Infections/virology
16.
J Virol ; 89(2): 1468-73, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25378499

ABSTRACT

Toll-like receptor 7 and Myd88 are required for antiretroviral antibody and germinal center responses, but whether somatic hypermutation and class-switch recombination are required for antiretroviral immunity has not been examined. Mice deficient in activation-induced cytidine deaminase (AID) resisted Friend virus infection, produced virus-neutralizing antibodies, and controlled viremia. Passive transfer demonstrated that immune IgM from AID-deficient mice contributes to Friend virus control in the presence of virus-specific CD4+ T cells.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Friend murine leukemia virus/immunology , Leukemia, Experimental/immunology , Retroviridae Infections/immunology , Somatic Hypermutation, Immunoglobulin , Tumor Virus Infections/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cytidine Deaminase/deficiency , Immunization, Passive , Immunoglobulin Class Switching , Immunoglobulin M/immunology , Leukemia, Experimental/virology , Mice , Retroviridae Infections/virology , Tumor Virus Infections/virology
17.
J Virol ; 89(1): 155-64, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25320301

ABSTRACT

UNLABELLED: Interleukin-1 beta (IL-1ß) is an inflammatory cytokine that is secreted in response to inflammasome activation by innate microbe-sensing pathways. Although some retroviruses can trigger IL-1ß secretion through the DNA-sensing molecule IFI16, the effect of IL-1ß on the course of infection is unknown. To test whether IL-1ß secretion affects retroviral replication in vivo, I constructed a novel murine leukemia virus strain (FMLV-IL-1ß) that encodes the mature form of IL-1ß. This virus replicated with kinetics similar to that of wild-type virus in tissue culture but caused a dramatically more aggressive infection of both C57BL/6 and BALB/c mice. By 7 days postinfection (PI), mice infected with FMLV-IL-1ß exhibited splenomegaly and viral loads 300-fold higher than those in mice infected with wild-type FMLV. Furthermore, the enlarged spleens of FMLV-IL-1ß-infected mice correlated with a large expansion of Gr-1(+) CD11b(+) myeloid-derived suppressor cells, as well as elevated levels of immune activation. Although FMLV-IL-1ß infection was controlled by C57BL/6 mice by 14 days p.i., FMLV-IL-1ß was able to establish a significant persistent infection and immune activation in BALB/c mice. These results demonstrate that IL-1ß secretion is a powerful positive regulator of retroviral infection and that FMLV-IL-1ß represents a new model of proinflammatory retroviral infection. IMPORTANCE: Interleukin-1 beta (IL-1ß) is an inflammatory cytokine released in response to activation of innate pathogen-sensing pathways during microbial infection. To examine the potential impact of IL-1ß on retroviral replication in vivo, I constructed a novel mouse retrovirus strain (FMLV-IL-1ß) that encodes IL-1ß and promotes abundant IL-1ß secretion from infected cells. This virus replicates with normal kinetics in cultured cells but displays a dramatically enhanced ability to replicate in mice and caused persistent infection and immune activation in the BALB/c strain of mice. These results establish IL-1ß as a positive regulator of retroviral replication and suggest that targeting this pathway may have therapeutic benefits in infections with proinflammatory retroviruses. This virus can also be used to further study the impact of inflammatory pathways on retroviral infection.


Subject(s)
Interleukin-1beta/metabolism , Leukemia Virus, Murine/immunology , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/pathology , Retroviridae Infections/pathology , Tumor Virus Infections/pathology , Animals , Host-Pathogen Interactions , Interleukin-1beta/genetics , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/physiology , Leukemia, Experimental/virology , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retroviridae Infections/virology , Splenomegaly/pathology , Tumor Virus Infections/virology , Viral Load , Virulence , Virus Replication
18.
Proc Biol Sci ; 282(1798): 20141568, 2015 Jan 07.
Article in English | MEDLINE | ID: mdl-25392466

ABSTRACT

Reduced genetic variation among hosts may favour the emergence of virulent infectious diseases by enhancing pathogen replication and its associated virulence due to adaptation to a limited set of host genotypes. Here, we test this hypothesis using experimental evolution of a mouse-specific retroviral pathogen, Friend virus (FV) complex. We demonstrate rapid fitness (i.e. viral titre) and virulence increases when FV complex serially infects a series of inbred mice representing the same genotype, but not when infecting a diverse array of inbred mouse strains modelling the diversity in natural host populations. Additionally, a single infection of a different host genotype was sufficient to constrain the emergence of a high fitness/high virulence FV complex phenotype in these experiments. The potent inhibition of viral fitness and virulence was associated with an observed loss of the defective retroviral genome (spleen focus-forming virus), whose presence exacerbates infection and drives disease in susceptible mice. Results from our experiments provide an important first step in understanding how genetic variation among vertebrate hosts influences pathogen evolution and suggests that serial exposure to different genotypes within a single host species may act as a constraint on pathogen adaptation that prohibits the emergence of more virulent infections. From a practical perspective, these results have implications for low-diversity host populations such as endangered species and domestic animals.


Subject(s)
Friend murine leukemia virus/physiology , Friend murine leukemia virus/pathogenicity , Genetic Fitness , Genotype , Host-Pathogen Interactions/genetics , Leukemia, Experimental/genetics , Retroviridae Infections/genetics , Tumor Virus Infections/genetics , Animals , Biological Evolution , Female , Leukemia, Experimental/virology , Mice , Mice, Inbred Strains , Retroviridae Infections/virology , Specific Pathogen-Free Organisms , Tumor Virus Infections/virology , Virulence/physiology
19.
Retrovirology ; 11: 126, 2014 Dec 24.
Article in English | MEDLINE | ID: mdl-25539593

ABSTRACT

BACKGROUND: Pathogen recognition drives host defense towards viral infections. Specific groups rather than single members of the protein family of pattern recognition receptors (PRRs) such as membrane spanning Toll-like receptors (TLRs) and cytosolic helicases might mediate sensing of replication intermediates of a specific virus species. TLR7 mediates host sensing of retroviruses and could significantly influence retrovirus-specific antibody responses. However, the origin of efficient cell-mediated immunity towards retroviruses is unknown. Double-stranded RNA intermediates produced during retroviral replication are good candidates for immune stimulatory viral products. Thus, we considered TLR3 as primer of cell-mediated immunity against retroviruses in vivo. RESULTS: Infection of mice deficient in TLR3 (TLR3(-/-)) with Friend retrovirus (FV) complex revealed higher viral loads during acute retroviral infection compared to wild type mice. TLR3(-/-) mice exhibited significantly lower expression levels of type I interferons (IFNs) and IFN-stimulated genes like Pkr or Ifi44, as well as reduced numbers of activated myeloid dendritic cells (DCs) (CD86(+) and MHC-II(+)). DCs generated from FV-infected TLR3(-/-) mice were less capable of priming virus-specific CD8(+) T cell proliferation. Moreover, cytotoxicity of natural killer (NK) cells as well as CD8(+) T cells were reduced in vitro and in vivo, respectively, in FV-infected TLR3(-/-) mice. CONCLUSIONS: TLR3 mediates antiretroviral cytotoxic NK cell and CD8(+) T cell activity in vivo. Our findings qualify TLR3 as target of immune therapy against retroviral infections.


Subject(s)
Friend murine leukemia virus/immunology , Toll-Like Receptor 3/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Killer Cells, Natural/immunology , Leukemia, Experimental/immunology , Leukemia, Experimental/virology , Mice, Inbred C57BL , Mice, Knockout , Retroviridae Infections/immunology , Retroviridae Infections/virology , Toll-Like Receptor 3/deficiency , Tumor Virus Infections/immunology , Tumor Virus Infections/virology , Viral Load
20.
Virology ; 468-470: 601-608, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25303118

ABSTRACT

APOBEC1 is a cytidine deaminase involved in cholesterol metabolism that has been linked to retrovirus restriction, analogous to the evolutionarily-related APOBEC3 proteins. In particular, murine APOBEC1 was shown to inhibit Friend retrovirus (FV) in vitro, generating high levels of C-to-T and G-to-A mutations. These observations raised the possibility that FV infection might be altered in APOBEC1-null mice. To examine this question directly, we infected wild-type and APOBEC1-null mice with FV complex and evaluated acute infection levels. Surprisingly, APOBEC1-null mice exhibited similar cellular infection levels and plasma viremia relative to wild-type mice. Moreover, next-generation sequencing analyses revealed that in contrast to APOBEC3, APOBEC1 did not enhance retroviral C-to-T and G-to-A mutational frequencies in genomic DNA. Thus, APOBEC1 neither inhibited nor significantly drove the molecular evolution of FV in vivo. Our findings reinforce that not all retrovirus restriction factors characterized as potent in vitro may be functionally relevant in vivo.


Subject(s)
Cytidine Deaminase/metabolism , Friend murine leukemia virus/physiology , Leukemia, Experimental/virology , Retroviridae Infections/virology , Tumor Virus Infections/virology , APOBEC-1 Deaminase , Animals , Cytidine Deaminase/genetics , Friend murine leukemia virus/genetics , Gene Expression Regulation/physiology , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mutation , Retroviridae Infections/genetics , Retroviridae Infections/metabolism , Tumor Virus Infections/genetics , Tumor Virus Infections/metabolism , Viral Load , Viremia , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL