Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 320
1.
Viruses ; 13(9)2021 08 27.
Article En | MEDLINE | ID: mdl-34578292

Small ruminant lentiviruses (SRLVs) infections lead to chronic diseases and remarkable economic losses undermining health and welfare of animals and the sustainability of farms. Early and definite diagnosis of SRLVs infections is the cornerstone for any control and eradication efforts; however, a "gold standard" test and/or diagnostic protocols with extensive applicability have yet to be developed. The main challenges preventing the development of a universally accepted diagnostic tool with sufficient sensitivity, specificity, and accuracy to be integrated in SRLVs control programs are the genetic variability of SRLVs associated with mutations, recombination, and cross-species transmission and the peculiarities of small ruminants' humoral immune response regarding late seroconversion, as well as intermittent and epitope-specific antibody production. The objectives of this review paper were to summarize the available serological and molecular assays for the diagnosis of SRLVs, to highlight their diagnostic performance emphasizing on advantages and drawbacks of their application, and to discuss current and future perspectives, challenges, limitations and impacts regarding the development of reliable and efficient tools for the diagnosis of SRLVs infections.


Lentivirus Infections/diagnosis , Lentivirus Infections/immunology , Lentivirus/genetics , Lentivirus/immunology , Ruminants/virology , Serologic Tests/veterinary , Animals , Arthritis-Encephalitis Virus, Caprine/genetics , Arthritis-Encephalitis Virus, Caprine/immunology , Goat Diseases/diagnosis , Goat Diseases/virology , Goats/virology , Lentivirus/classification , Lentivirus/isolation & purification , Seroconversion , Serologic Tests/methods , Sheep/virology , Sheep Diseases/diagnosis , Sheep Diseases/virology , Virology/methods , Visna-maedi virus/genetics , Visna-maedi virus/immunology
2.
Viruses ; 13(7)2021 07 01.
Article En | MEDLINE | ID: mdl-34372496

Maedi-visna virus (MVV) and caprine arthritis encephalitis virus (CAEV), referred to as small ruminant lentiviruses (SRLVs), belong to the genus Lentivirus of the Retroviridae family. SRLVs infect both sheep and goats, causing significant economic losses and animal welfare damage. Recent findings suggest an association between serological status and allelic variants of different genes such as TMEM154, TLR9, MYD88 and CCR5. The aim of this work was to investigate the role of specific polymorphisms of these genes in SRLVs infection in some sheep flocks in Italy. In addition to those already known, novel variants in the TMEM154 (P7H, I74V, I105V) gene were detected in this study. The risk of infection was determined finding an association between the serological status and polymorphisms P7H, E35K, N70I, I74V, I105V of TMEM154, R447Q, A462S and G520R in TLR9 gene, H176H* and K190K* in MYD88 genes, while no statistical association was observed for the 4-bp deletion of the CCR5 gene. Since no vaccines or treatments have been developed, a genetically based approach could be an innovative strategy to prevent and to control SRLVs infection. Our findings are an important starting point in order to define the genetic resistance profile towards SRLVs infection.


Disease Resistance/genetics , Lentivirus Infections/genetics , Lentivirus Infections/veterinary , Lentivirus/genetics , Membrane Proteins/genetics , Myeloid Differentiation Factor 88/genetics , Polymorphism, Single Nucleotide , Receptors, CCR5/genetics , Toll-Like Receptor 9/genetics , Animals , Genetic Variation , Italy , Lentivirus/classification , Lentivirus Infections/immunology , Lentivirus Infections/prevention & control , Membrane Proteins/classification , Membrane Proteins/immunology , Risk Factors , Sheep , Sheep Diseases/genetics , Sheep Diseases/immunology , Sheep Diseases/virology
3.
Front Immunol ; 12: 638907, 2021.
Article En | MEDLINE | ID: mdl-33953713

Chimeric antigen receptor (CAR)-T cell therapy has become an important method for the treatment of hematological tumors. Lentiviruses are commonly used gene transfer vectors for preparing CAR-T cells, and the conditions for preparing CAR-T cells vary greatly. This study reported for the first time the influence of differences in infection temperature on the phenotype and function of produced CAR-T cells. Our results show that infection at 4 degrees produces the highest CAR-positive rate of T cells, infection at 37 degrees produces the fastest proliferation in CAR-T cells, and infection at 32 degrees produces CAR-T cells with the greatest proportion of naive cells and the lowest expression of immune checkpoints. Therefore, infection at 32 degrees is recommended to prepare CAR-T cells. CAR-T cells derived from infection at 32 degrees seem to have a balance between function and phenotype. Importantly, they have increased oncolytic ability. This research will help optimize the generation of CAR-T cells and improve the quality of CAR-T cell products.


Genetic Vectors/genetics , Hematologic Neoplasms/therapy , Immunotherapy, Adoptive/methods , Lentivirus Infections/immunology , Lentivirus/genetics , Animals , Cytotoxicity, Immunologic , Disease Models, Animal , HEK293 Cells , Heterografts , Humans , Male , Mice , Phenotype , Temperature
4.
Immunology ; 164(2): 348-357, 2021 10.
Article En | MEDLINE | ID: mdl-34037988

Granulocytes mediate broad immunoprotection through phagocytosis, extracellular traps, release of cytotoxic granules, antibody effector functions and recruitment of other immune cells against pathogens. However, descriptions of granulocytes in HIV infection and mucosal tissues are limited. Our goal was to characterize granulocyte subsets in systemic, mucosal and lymphoid tissues during lentiviral infection using the rhesus macaque (RM) model. Mononuclear cells from jejunum, colon, cervix, vagina, lymph nodes, spleen, liver and whole blood from experimentally naïve and chronically SHIVsf162p3-infected RM were analysed by microscopy and polychromatic flow cytometry. Granulocytes were identified using phenotypes designed specifically for RM: eosinophils-CD45+  CD66+  CD49d+ ; neutrophils-CD45+  CD66+  CD14+ ; and basophils-CD45+  CD123+  FcRε+ . Nuclear visualization with DAPI staining and surface marker images by ImageStream (cytometry/microscopy) further confirmed granulocytic phenotypes. Flow cytometric data showed that all RM granulocytes expressed CD32 (FcRγII) but did not express CD16 (FcRγIII). Additionally, constitutive expression of CD64 (FcRγI) on neutrophils and FcRε on basophils indicates the differential expression of Fc receptors on granulocyte subsets. Granulocytic subsets in naïve whole blood ranged from 25·4% to 81·5% neutrophils, 0·59% to 13·3% eosinophils and 0·059% to 1·8% basophils. Interestingly, elevated frequencies of circulating neutrophils, colorectal neutrophils and colorectal eosinophils were all observed in chronic lentiviral disease. Conversely, circulating basophils, jejunal eosinophils, vaginal neutrophils and vaginal eosinophils of SHIVsf162p3-infected RM declined in frequency. Overall, our data suggest modulation of granulocytes in chronic lentiviral infection, most notably in the gastrointestinal mucosae where a significant inflammation and disruption occurs in lentivirus-induced disease. Furthermore, granulocytes may migrate to inflamed tissues during infection and could serve as targets of immunotherapeutic intervention.


Granulocytes/immunology , Lentivirus Infections/immunology , Macaca mulatta/immunology , Mucous Membrane/immunology , Animals , Basophils/immunology , Basophils/virology , Eosinophils/immunology , Eosinophils/virology , Flow Cytometry/methods , Granulocytes/virology , HIV Infections/immunology , HIV Infections/virology , Leukocyte Count/methods , Mucous Membrane/virology , Neutrophils/immunology , Neutrophils/virology , Receptors, IgG/immunology
5.
EBioMedicine ; 60: 103008, 2020 Oct.
Article En | MEDLINE | ID: mdl-32979832

BACKGROUND: Memory CD8+T cell responses play an essential role in protection against persistent infection. However, HIV-1 evades vaccine-induced memory CD8+T cell response by mechanisms that are not fully understood. METHODS: We analyzed the temporal dynamics of CD8+T cell recall activity and function during EcoHIV infection in a potent PD1-based vaccine immunized immunocompetent mice. FINDINGS: Upon intraperitoneal EcoHIV infection, high levels of HIV-1 GAG-specific CD8+T lymphocytes recall response reduced EcoHIV-infected cells significantly. However, this protective effect diminished quickly after seven days, followed by a rapid reduction of GAG-specific CD8+T cell number and activity, and viral persistence. Mechanistically, EcoHIV activated dendritic cells (DCs) and myeloid cells. Myeloid cells were infected and rapidly expanded, exhibiting elevated PD-L1/-L2 expression and T cell suppressive function before day 7, and were resistant to CD8+T cell-mediated apoptosis. Depletion of myeloid-derived suppressor cells (MDSCs) reduced EcoHIV infection and boosted T cell responses. INTERPRETATION: This study provides an overview of the temporal interplay of persistent virus, DCs, MDSCs and antigen-specific CD8+T cells during acute infection. We identify MDSCs as critical gatekeepers that restrain antiviral T cell memory responses, and highlight MDSCs as an important target for developing effective vaccines against chronic human infections. FUNDING: Hong Kong Research Grant Council (T11-709/18-N, HKU5/CRF/13G), General Research Fund (17122915 and 17114114), Hong Kong Health and Medical Research Fund (11100752, 14130582, 16150662), Grant RGC-ANR A-HKU709/14, the San-Ming Project of Medicine (SZSM201512029), University Development Fund of the University of Hong Kong and Li Ka Shing Faculty of Medicine Matching Fund to HKU AIDS Institute.


CD8-Positive T-Lymphocytes/immunology , Host-Pathogen Interactions/immunology , Immunologic Memory , Lentivirus Infections/immunology , Lentivirus Infections/virology , Lentivirus/immunology , Myeloid-Derived Suppressor Cells/immunology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Biomarkers , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , HIV Infections/immunology , HIV Infections/metabolism , HIV Infections/virology , HIV-1/genetics , HIV-1/immunology , Humans , Immunocompetence , Immunomodulation , Lentivirus/genetics , Lentivirus Infections/metabolism , Lymphocyte Activation/immunology , Lymphocyte Depletion , Mice , Mice, Transgenic , Myeloid-Derived Suppressor Cells/metabolism , Viral Load , Viral Vaccines/immunology
6.
Arch Virol ; 165(7): 1557-1567, 2020 Jul.
Article En | MEDLINE | ID: mdl-32356187

Caprine arthritis-encephalitis (CAE) is a chronic progressive infectious disease caused by caprine arthritis-encephalitis virus (CAEV) that seriously threatens the goat industry. Chronic infection and life-long multi-tissue inflammation are the typical features of the disease. Innate antiviral immunity is essential for the host defense system that rapidly recognizes and eliminates invading viruses. Interferon ß (IFN-ß) is important for innate immunity and regulates immunity against a broad spectrum of viruses. To investigate the details of the IFN-ß response to CAEV infection, the effects of six viral proteins and the molecular mechanisms by which they affect IFN-ß production were analyzed. Overexpression of DU and Vif promote virus proliferation and inhibit the production of IFN-ß. qRT-PCR and luciferase reporter assays showed that overexpression of Vif inhibits the expression of luciferase under the control of the ISRE, NF-κB or IFN-ß promoter but does not affect the expression of IFN-ß activated by IRF3, indicating that Vif negatively regulates IFN-ß production by affecting upstream signal transduction of IRF3. Amino acids 149-164 of Vif were found to be necessary for the inhibitory effect of IFN-ß production. Our results indicate that CAEV evades surveillance and clearance by intracellular innate immunity by downregulating IFN-ß production.


Arthritis-Encephalitis Virus, Caprine/immunology , Gene Products, vif/immunology , Goat Diseases/immunology , Interferon-beta/immunology , Lentivirus Infections/veterinary , Animals , Arthritis-Encephalitis Virus, Caprine/genetics , Gene Products, vif/genetics , Goat Diseases/genetics , Goat Diseases/virology , Goats , Host-Pathogen Interactions , Immunity, Innate , Interferon-beta/genetics , Lentivirus Infections/genetics , Lentivirus Infections/immunology , Lentivirus Infections/virology , NF-kappa B/genetics , NF-kappa B/immunology
7.
Viruses ; 12(6)2020 05 27.
Article En | MEDLINE | ID: mdl-32471198

Mammals have developed clever adaptive and innate immune defense mechanisms to protect against invading bacterial and viral pathogens. Human innate immunity is continuously evolving to expand the repertoire of restriction factors and one such family of intrinsic restriction factors is the APOBEC3 (A3) family of cytidine deaminases. The coordinated expression of seven members of the A3 family of cytidine deaminases provides intrinsic immunity against numerous foreign infectious agents and protects the host from exogenous retroviruses and endogenous retroelements. Four members of the A3 proteins-A3G, A3F, A3H, and A3D-restrict HIV-1 in the absence of virion infectivity factor (Vif); their incorporation into progeny virions is a prerequisite for cytidine deaminase-dependent and -independent activities that inhibit viral replication in the host target cell. HIV-1 encodes Vif, an accessory protein that antagonizes A3 proteins by targeting them for polyubiquitination and subsequent proteasomal degradation in the virus producing cells. In this review, we summarize our current understanding of the role of human A3 proteins as barriers against HIV-1 infection, how Vif overcomes their antiviral activity, and highlight recent structural and functional insights into A3-mediated restriction of lentiviruses.


APOBEC Deaminases/physiology , Lentivirus Infections/immunology , Lentivirus/immunology , APOBEC Deaminases/chemistry , Animals , Base Sequence , Humans , Lentivirus/metabolism , Lentivirus Infections/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
8.
Viruses ; 11(9)2019 09 07.
Article En | MEDLINE | ID: mdl-31500260

Feline immunodeficiency virus (FIV) is a naturally occurring T-cell tropic lentiviral disease of felids with many similarities to HIV/AIDS in humans. Similar to primate lentiviral-host interactions, feline APOBEC3 (A3) has been shown to inhibit FIV infection in a host-specific manner and feline A3 degradation is mediated by FIV Vif. Further, infection of felids with non-native FIV strains results in restricted viral replication in both experimental and naturally occurring infections. However, the link between molecular A3-Vif interactions and A3 biological activity during FIV infection has not been well characterized. We thus examined expression of the feline A3 genes A3Z2, A3Z3 and A3Z2-Z3 during experimental infection of domestic cats with host-adapted domestic cat FIV (referred to as FIV) and non-adapted Puma concolor FIV (referred to as puma lentivirus, PLV). We determined A3 expression in different tissues and blood cells from uninfected, FIV-infected, PLV-infected and FIV/PLV co-infected cats; and in purified blood cell subpopulations from FIV-infected and uninfected cats. Additionally, we evaluated regulation of A3 expression by cytokines, mitogens, and FIV infection in cultured cells. In all feline cells and tissues studied, there was a striking difference in expression between the A3 genes which encode FIV inhibitors, with A3Z3 mRNA abundance exceeding that of A3Z2-Z3 by 300-fold or more. Interferon-alpha treatment of cat T cells resulted in upregulation of A3 expression, while treatment with interferon-gamma enhanced expression in cat cell lines. In cats, secondary lymphoid organs and peripheral blood mononuclear cells (PBMC) had the highest basal A3 expression levels and A3 genes were differentially expressed among blood T cells, B cells, and monocytes. Acute FIV and PLV infection of cats, and FIV infection of primary PBMC resulted in no detectable change in A3 expression with the exception of significantly elevated A3 expression in the thymus, the site of highest FIV replication. We conclude that cat A3 expression is regulated by cytokine treatment but, by and large, lentiviral infection did not appear to alter expression. Differences in A3 expression in different blood cell subsets did not appear to impact FIV viral replication kinetics within these cells. Furthermore, the relative abundance of A3Z3 mRNA compared to A3Z2-Z3 suggests that A3Z3 may be the major active anti-lentiviral APOBEC3 gene product in domestic cats.


Cytosine Deaminase/immunology , Feline Acquired Immunodeficiency Syndrome/enzymology , Immunodeficiency Virus, Feline/physiology , Lentivirus Infections/veterinary , Animals , B-Lymphocytes/immunology , Cats , Cytosine Deaminase/genetics , Feline Acquired Immunodeficiency Syndrome/genetics , Feline Acquired Immunodeficiency Syndrome/immunology , Feline Acquired Immunodeficiency Syndrome/virology , Host-Pathogen Interactions , Immunodeficiency Virus, Feline/genetics , Lentivirus Infections/enzymology , Lentivirus Infections/genetics , Lentivirus Infections/immunology , T-Lymphocytes/immunology , Virus Replication
9.
Fish Shellfish Immunol ; 86: 1114-1122, 2019 Mar.
Article En | MEDLINE | ID: mdl-30594581

RIG-I-like receptors (RLRs), as key cytoplasmic sensors of viral pathogen-associated molecular patterns, can recognise viral RNA and enhance the antiviral response. Some investigations have focused on the roles of RLRs in the innate immune response in grass carp, large yellow croaker, and rainbow trout. However, little is known about the function of RLRs in mandarinfish (Siniperca chuatsi), an important economic fish in Perciformes. Here, we functionally characterized the RLRs involved in the immune responses of mandarinfish (Siniperca chuatsi), by evaluating three RLRs, namely, RIG-I, MDA5, and LGP2. The results revealed that MDA5 and LGP2 were present in mandarinfish, whereas RIG-I was absent. The MDA5 and LGP2 cDNA sequences contained 2976 and 2046 bp and encoded 991 and 681 amino acids, respectively. Multiple sequence alignments showed that MDA5 and LGP2 of mandarinfish were clustered together with their homologs from other teleost fishes and shared high similarities with those from other vertebrates, and RIG-I of mandarinfish was absent. Moreover, quantitative real-time PCR (qPCR) analysis suggested that MDA5 and LGP2 were constitutively expressed in all tissues tested, and MDA5 mRNA expression was relatively high in the gill, and spleen, whereas LGP2 mRNA expression was high in the liver, gill, and head kidney. After stimulation with lipopolysaccharide or poly I:C, the expression of MDA5 and LGP2 was upregulated in spleen, gill and head kidney, but the pattern was not exactly the same, MDA5 transcripts generally increased and then declined with the prolonged infection, while LGP2 transcripts went up continuously, which showed that mandarinfish MDA5 and LGP2 may play independent roles in antiviral response. Besides, it is further revealed that the MDA5 could activate NF-κB and IRF3 to inducing the production of IFN-ß by constructing tet-on stable strain of 293T cell, however over-expression of LGP2 resulted in decreased NF-κB, IRF3 and IFN-ß production in cells challenged with LPS and polyI:C Taken together, our results demonstrated that MDA5 and LGP2, as a positive and negative regulator, respectively, played an important role in modulating antibacterial andantiviral immune responses though activating NF-κB and IRF3 in RLRs signaling of mandarinfish.


Fish Proteins/metabolism , Interferon Regulatory Factors/metabolism , Interferon-Induced Helicase, IFIH1/metabolism , NF-kappa B/metabolism , Perciformes/metabolism , RNA Helicases/metabolism , Animals , Fish Diseases/immunology , Fish Diseases/virology , Fish Proteins/genetics , Gene Expression Regulation , HEK293 Cells , Humans , Immunity, Innate , Interferon-Induced Helicase, IFIH1/genetics , Lentivirus , Lentivirus Infections/immunology , Lipopolysaccharides/pharmacology , Poly I-C/immunology , RNA Helicases/genetics , Sequence Alignment , Sequence Analysis, Protein , Signal Transduction
10.
Nat Commun ; 9(1): 5193, 2018 12 05.
Article En | MEDLINE | ID: mdl-30518925

Immunosuppression is a hallmark of tumor progression, and treatments that inhibit or deplete monocytic myeloid-derived suppressive cells could promote anti-tumor immunity. c-FLIP is a central regulator of caspase-8-mediated apoptosis and necroptosis. Here we show that low-dose cytotoxic chemotherapy agents cause apoptosis linked to c-FLIP down-regulation selectively in monocytes. Enforced expression of c-FLIP or viral FLIP rescues monocytes from cytotoxicity and concurrently induces potent immunosuppressive activity, in T cell cultures and in vivo models of tumor progression and immunotherapy. FLIP-transduced human blood monocytes can suppress graft versus host disease. Neither expression of FLIP in granulocytes nor expression of other anti-apoptotic genes in monocytes conferred immunosuppression, suggesting that FLIP effects on immunosuppression are specific to monocytic lineage and distinct from death inhibition. Mechanistically, FLIP controls a broad transcriptional program, partially by NF-κB activation. Therefore, modulation of FLIP in monocytes offers a means to elicit or block immunosuppressive myeloid cells.


CASP8 and FADD-Like Apoptosis Regulating Protein/immunology , Lentivirus Infections/immunology , Monocytes/immunology , NF-kappa B/immunology , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cells, Cultured , Humans , Immunosuppression Therapy , Lentivirus/physiology , Lentivirus Infections/genetics , Lentivirus Infections/physiopathology , Lentivirus Infections/virology , Myeloid Cells/immunology , NF-kappa B/genetics
11.
Viruses ; 10(8)2018 08 17.
Article En | MEDLINE | ID: mdl-30126090

Lentiviruses are infectious agents of a number of animal species, including sheep, goats, horses, monkeys, cows, and cats, in addition to humans. As in the human case, the host immune response fails to control the establishment of chronic persistent infection that finally leads to a specific disease development. Despite intensive research on the development of lentivirus vaccines, it is still not clear which immune responses can protect against infection. Viral mutations resulting in escape from T-cell or antibody-mediated responses are the basis of the immune failure to control the infection. The innate immune response provides the first line of defense against viral infections in an antigen-independent manner. Antiviral innate responses are conducted by dendritic cells, macrophages, and natural killer cells, often targeted by lentiviruses, and intrinsic antiviral mechanisms exerted by all cells. Intrinsic responses depend on the recognition of the viral pathogen-associated molecular patterns (PAMPs) by pathogen recognition receptors (PRRs), and the signaling cascades leading to an antiviral state by inducing the expression of antiviral proteins, including restriction factors. This review describes the latest advances on innate immunity related to the infection by animal lentiviruses, centered on small ruminant lentiviruses (SRLV), equine infectious anemia virus (EIAV), and feline (FIV) and bovine immunodeficiency viruses (BIV), specifically focusing on the antiviral role of the major restriction factors described thus far.


Gene Expression Regulation/immunology , Immunity, Innate , Interferon Regulatory Factors/immunology , Lentivirus Infections/immunology , Receptors, Pattern Recognition/immunology , Animals , Cats , Cattle , Dendritic Cells/immunology , Dendritic Cells/virology , Goats , Horses , Immunodeficiency Virus, Bovine/immunology , Immunodeficiency Virus, Bovine/pathogenicity , Immunodeficiency Virus, Feline/immunology , Immunodeficiency Virus, Feline/pathogenicity , Infectious Anemia Virus, Equine/immunology , Infectious Anemia Virus, Equine/pathogenicity , Interferon Regulatory Factors/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Lentivirus Infections/genetics , Lentivirus Infections/virology , Macrophages/immunology , Macrophages/virology , Pathogen-Associated Molecular Pattern Molecules/immunology , Receptors, Pattern Recognition/genetics , Sheep , T-Lymphocytes/immunology , T-Lymphocytes/virology
12.
Anim Sci J ; 89(9): 1364-1370, 2018 Sep.
Article En | MEDLINE | ID: mdl-29877006

We carried out this study to determine for how long small ruminant lentivirus (SRLV)-specific antibodies can be detected by three commercial ELISA kits in goat kids after suckling infected does in field conditions. Forty-one kids born to SRLV-seropositive asymptomatic does were blood sampled prior to colostrum consumption, and then weekly for 6 months in total. The sera were screened with three commercial ELISA kits: whole-virus ELISA (wELISA), recombinant transmembrane and capsid antigen ELISA (TM/CA-ELISA), and surface antigen ELISA (SU-ELISA). All but one kid were seronegative in all three ELISAs right after birth. At the age of 1 week all kids turned seropositive in wELISA, 39 kids (95%) in TM/CA-ELISA, and 35 kids (85%) in SU-ELISA. All seropositive kids turned seronegative in wELISA by the 15th week, and in SU-ELISA by the 19th week (median of 8 weeks in both ELISA), whereas in TM/CA-ELISA five kids (13% of 39 initially seropositive) were still seropositive at the age of 6 months (median of 11 weeks). Antibody levels at the age of 1 week proved significantly linked to the duration of maternal antibodies in all three ELISAs and could be employed to predict for how long maternal antibodies would remain detectable.


Animals, Newborn/immunology , Antibodies, Viral/blood , Goat Diseases/immunology , Goat Diseases/virology , Lentivirus Infections/immunology , Lentivirus Infections/veterinary , Lentivirus/immunology , Maternal-Fetal Exchange , Animals , Enzyme-Linked Immunosorbent Assay/methods , Female , Goats , Lentivirus Infections/virology , Male , Pregnancy , Time Factors
13.
Viruses ; 10(5)2018 05 02.
Article En | MEDLINE | ID: mdl-29724026

(1) Background: Small ruminant lentiviruses (SRLV) persist in infected goats that mount a strong humoral immune response characterized by low neutralizing titers. In this study, we characterized the antibody response to SU5, a variable, immunodominant epitope of the envelope glycoprotein of SRLV. We tested the working hypothesis that the variability of SU5 reflects escape from neutralizing antibody. (2) Methods: Affinity purified anti-SU5 antibody were tested for their neutralizing activity to the homologous lentivirus. Virus culture supernatant—in native form or following sonication and filtration—was used to test the ability of free envelope glycoproteins to compete for binding in a SU5-peptide-ELISA. (3) Results: Anti-SU5 antibodies are not neutralizing, strongly suggesting that they do not bind intact viral particles. In contrast, shed envelope glycoproteins efficiently compete for binding in a SU5-ELISA, providing convincing evidence that the SU5 epitope is exposed only on shed envelope glycoproteins. (4) Conclusions: Our results show that the antibody engaging SU5 is not neutralizing and does not appear to bind to SU expressed at the surface of virus particles. We propose that SU5 is a potential decoy epitope exposed on shaded envelope glycoproteins, luring the humoral immune response in committing an original antigenic sin to a functionally irrelevant epitope.


Antibodies, Neutralizing/immunology , Antigens, Viral/immunology , Arthritis-Encephalitis Virus, Caprine/immunology , Immunodominant Epitopes/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Viral/immunology , Antigens, Viral/genetics , Enzyme-Linked Immunosorbent Assay , Goat Diseases/immunology , Goat Diseases/virology , Goats/immunology , Goats/virology , Immunodominant Epitopes/genetics , Lentivirus Infections/immunology , Lentivirus Infections/veterinary , Neutralization Tests , Peptides/immunology , Viral Envelope Proteins/genetics
14.
Viruses ; 10(5)2018 04 28.
Article En | MEDLINE | ID: mdl-29710792

CD8⁺ T cells are critical for controlling viremia during human immunodeficiency virus (HIV) infection. These cells produce cytolytic factors and antiviral cytokines that eliminate virally- infected cells. During the chronic phase of HIV infection, CD8⁺ T cells progressively lose their proliferative capacity and antiviral functions. These dysfunctional cells are unable to clear the productively infected and reactivated cells, representing a roadblock in HIV cure. Therefore, mechanisms to understand CD8⁺ T cell dysfunction and strategies to boost CD8⁺ T cell function need to be investigated. Using the feline immunodeficiency virus (FIV) model for lentiviral persistence, we have demonstrated that CD8⁺ T cells exhibit epigenetic changes such as DNA demethylation during the course of infection as compared to uninfected cats. We have also demonstrated that lentivirus-activated CD4⁺CD25⁺ T regulatory cells induce forkhead box P3 (Foxp3) expression in virus-specific CD8⁺ T cell targets, which binds the interleukin (IL)-2, tumor necrosis factor (TNF)-α, and interferon (IFN)-γ promoters in these CD8⁺ T cells. Finally, we have reported that epigenetic modulation reduces Foxp3 binding to these promoter regions. This review compares and contrasts our current understanding of CD8⁺ T cell epigenetics and mechanisms of lymphocyte suppression during the course of lentiviral infection for two animal models, FIV and simian immunodeficiency virus (SIV).


CD8-Positive T-Lymphocytes/immunology , Epigenesis, Genetic , Immunodeficiency Virus, Feline , Lentivirus Infections/immunology , Lentivirus Infections/veterinary , Simian Immunodeficiency Virus , Animals , Cats , Cytokines/immunology , HIV Infections/immunology , Humans , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Viremia/veterinary
15.
Vet Immunol Immunopathol ; 188: 21-26, 2017 Jun.
Article En | MEDLINE | ID: mdl-28615124

The exact influence of caprine arthritis encephalitis virus (CAEV) infection on blood and milk polymorphonuclear leukocytes (PMNLs) and monocyte/macrophages of goats remains unclear. Thus, the present study sought to explore the blood and milk PMNL and monocyte/macrophage functions in naturally CAEV-infected goats. The present study used 18 healthy Saanen goats that were segregated according to sera test outcomes into serologically CAEV negative (n=8; 14 halves) and positive (n=10; 14 halves) groups. All milk samples from mammary halves with milk bacteriologically positive outcomes, somatic cell count ≥2×106cellsmL-1, and abnormal secretions in the strip cup test were excluded. We evaluated the percentage of blood and milk PMNLs and monocyte/macrophages, the viability of PMNLs and monocyte/macrophages, the levels of intracellular reactive oxygen species (ROS) and the nonopsonized phagocytosis of Staphylococcus aureus and Escherichia coli by flow cytometry. In the present study, a higher percentage of milk macrophages (CD14+) and milk polymorphonuclear leukocytes undergoing late apoptosis or necrosis (Annexin-V+/Propidium iodide+) was observed in CAEV-infected goats; we did not find any further alterations in blood and milk PMNL and monocyte/macrophage functions. Thus, regarding our results, the goats naturally infected with CAEV did not reveal pronounced dysfunctions in blood and milk polymorphonuclear leukocytes and monocytes/macrophages.


Arthritis-Encephalitis Virus, Caprine/immunology , Goat Diseases/virology , Lentivirus Infections/veterinary , Macrophages/immunology , Monocytes/immunology , Neutrophils/immunology , Animals , Female , Goat Diseases/blood , Goat Diseases/immunology , Goats/blood , Goats/immunology , Goats/virology , Lentivirus Infections/blood , Lentivirus Infections/immunology , Milk/cytology , Milk/immunology , Milk/virology
16.
JCI Insight ; 2(4): e91214, 2017 02 23.
Article En | MEDLINE | ID: mdl-28239657

SIV DNA can be detected in lymphoid tissue-resident macrophages of chronically SIV-infected Asian macaques. These macrophages also contain evidence of recently phagocytosed SIV-infected CD4+ T cells. Here, we examine whether these macrophages contain replication-competent virus, whether viral DNA can be detected in tissue-resident macrophages from antiretroviral (ARV) therapy-treated animals and humans, and how the viral sequences amplified from macrophages and contemporaneous CD4+ T cells compare. In ARV-naive animals, we find that lymphoid tissue-resident macrophages contain replication-competent virus if they also contain viral DNA in ARV-naive Asian macaques. The genetic sequence of the virus within these macrophages is similar to those within CD4+ T cells from the same anatomic sites. In ARV-treated animals, we find that viral DNA can be amplified from lymphoid tissue-resident macrophages of SIV-infected Asian macaques that were treated with ARVs for at least 5 months, but we could not detect replication-competent virus from macrophages of animals treated with ARVs. Finally, we could not detect viral DNA in alveolar macrophages from HIV-infected individuals who received ARVs for 3 years and had undetectable viral loads. These data demonstrate that macrophages can contain replication-competent virus, but may not represent a significant reservoir for HIV in vivo.


DNA, Viral/metabolism , HIV Infections/virology , HIV-1/genetics , Macrophages/virology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Virus Replication , Animals , Anti-Retroviral Agents/therapeutic use , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , HIV Infections/drug therapy , HIV Infections/immunology , HIV-1/physiology , Humans , Lentivirus Infections/drug therapy , Lentivirus Infections/immunology , Lentivirus Infections/virology , Lymphoid Tissue/cytology , Macaca , Macaca mulatta , Macaca nemestrina , Macrophages/immunology , Macrophages/metabolism , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/physiology , Viral Load
17.
JCI Insight ; 2(4): e91230, 2017 02 23.
Article En | MEDLINE | ID: mdl-28239658

The conditioning regimen used as part of the Berlin patient's hematopoietic cell transplant likely contributed to his eradication of HIV infection. We studied the impact of conditioning in simian-human immunodeficiency virus-infected (SHIV-infected) macaques suppressed by combination antiretroviral therapy (cART). The conditioning regimen resulted in a dramatic, but incomplete depletion of CD4+ and CD8+ T cells and CD20+ B cells, increased T cell activation and exhaustion, and a significant loss of SHIV-specific Abs. The disrupted T cell homeostasis and markers of microbial translocation positively correlated with an increased viral rebound after cART interruption. Quantitative viral outgrowth and Tat/rev-induced limiting dilution assays showed that the size of the latent SHIV reservoir did not correlate with viral rebound. These findings identify perturbations of the immune system as a mechanism for the failure of autologous transplantation to eradicate HIV. Thus, transplantation strategies may be improved by incorporating immune modulators to prevent disrupted homeostasis, and gene therapy to protect transplanted cells.


CD4-Positive T-Lymphocytes/radiation effects , CD8-Positive T-Lymphocytes/radiation effects , HIV Infections/immunology , HIV-1/radiation effects , Hematopoietic Stem Cell Transplantation/methods , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/radiation effects , Transplantation Conditioning/methods , Whole-Body Irradiation , Animals , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , HIV Infections/drug therapy , Homeostasis/radiation effects , Lentivirus Infections/drug therapy , Lentivirus Infections/immunology , Macaca nemestrina , Simian Acquired Immunodeficiency Syndrome/drug therapy , Transplantation, Autologous , Viral Load/radiation effects
18.
Immunity ; 45(2): 255-66, 2016 08 16.
Article En | MEDLINE | ID: mdl-27496731

Detection of intracellular DNA triggers activation of the STING-dependent interferon-stimulatory DNA (ISD) pathway, which is essential for antiviral responses. Multiple DNA sensors have been proposed to activate this pathway, including AIM2-like receptors (ALRs). Whether the ALRs are essential for activation of this pathway remains unknown. To rigorously explore the function of ALRs, we generated mice lacking all 13 ALR genes. We found that ALRs are dispensable for the type I interferon (IFN) response to transfected DNA ligands, DNA virus infection, and lentivirus infection. We also found that ALRs do not contribute to autoimmune disease in the Trex1(-/-) mouse model of Aicardi-Goutières Syndrome. Finally, CRISPR-mediated disruption of the human AIM2-like receptor IFI16 in primary fibroblasts revealed that IFI16 is not essential for the IFN response to human cytomegalovirus infection. Our findings indicate that ALRs are dispensable for the ISD response and suggest that alternative functions for these receptors should be explored.


Autoimmune Diseases of the Nervous System/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , DNA-Binding Proteins/metabolism , Lentivirus Infections/immunology , Lentivirus/immunology , Nervous System Malformations/immunology , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats , DNA/immunology , DNA-Binding Proteins/genetics , Disease Models, Animal , Exodeoxyribonucleases/genetics , Genetic Loci/genetics , Humans , Interferon Type I/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Phosphoproteins/genetics
19.
PLoS Pathog ; 11(9): e1005149, 2015 Sep.
Article En | MEDLINE | ID: mdl-26394054

Cross-species transmissions of viruses from animals to humans are at the origin of major human pathogenic viruses. While the role of ecological and epidemiological factors in the emergence of new pathogens is well documented, the importance of host factors is often unknown. Chimpanzees are the closest relatives of humans and the animal reservoir at the origin of the human AIDS pandemic. However, despite being regularly exposed to monkey lentiviruses through hunting, chimpanzees are naturally infected by only a single simian immunodeficiency virus, SIVcpz. Here, we asked why chimpanzees appear to be protected against the successful emergence of other SIVs. In particular, we investigated the role of the chimpanzee APOBEC3 genes in providing a barrier to infection by most monkey lentiviruses. We found that most SIV Vifs, including Vif from SIVwrc infecting western-red colobus, the chimpanzee's main monkey prey in West Africa, could not antagonize chimpanzee APOBEC3G. Moreover, chimpanzee APOBEC3D, as well as APOBEC3F and APOBEC3H, provided additional protection against SIV Vif antagonism. Consequently, lentiviral replication in primary chimpanzee CD4(+) T cells was dependent on the presence of a lentiviral vif gene that could antagonize chimpanzee APOBEC3s. Finally, by identifying and functionally characterizing several APOBEC3 gene polymorphisms in both common chimpanzees and bonobos, we found that these ape populations encode APOBEC3 proteins that are uniformly resistant to antagonism by monkey lentiviruses.


Cytidine Deaminase/genetics , Lentivirus Infections/genetics , Pan troglodytes/immunology , Pan troglodytes/virology , Simian Immunodeficiency Virus/genetics , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/immunology , Cytidine Deaminase/immunology , Genes, vif/genetics , Haplorhini , Lentivirus/genetics , Lentivirus Infections/immunology , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction
20.
PLoS Pathog ; 11(9): e1005146, 2015 Sep.
Article En | MEDLINE | ID: mdl-26360709

The virus-host relationship in simian immunodeficiency virus (SIV) infected chimpanzees is thought to be different from that found in other SIV infected African primates. However, studies of captive SIVcpz infected chimpanzees are limited. Previously, the natural SIVcpz infection of one chimpanzee, and the experimental infection of six chimpanzees was reported, with limited follow-up. Here, we present a long-term study of these seven animals, with a retrospective re-examination of the early stages of infection. The only clinical signs consistent with AIDS or AIDS associated disease was thrombocytopenia in two cases, associated with the development of anti-platelet antibodies. However, compared to uninfected and HIV-1 infected animals, SIVcpz infected animals had significantly lower levels of peripheral blood CD4+ T-cells. Despite this, levels of T-cell activation in chronic infection were not significantly elevated. In addition, while plasma levels of ß2 microglobulin, neopterin and soluble TNF-related apoptosis inducing ligand (sTRAIL) were elevated in acute infection, these markers returned to near-normal levels in chronic infection, reminiscent of immune activation patterns in 'natural host' species. Furthermore, plasma soluble CD14 was not elevated in chronic infection. However, examination of the secondary lymphoid environment revealed persistent changes to the lymphoid structure, including follicular hyperplasia in SIVcpz infected animals. In addition, both SIV and HIV-1 infected chimpanzees showed increased levels of deposition of collagen and increased levels of Mx1 expression in the T-cell zones of the lymph node. The outcome of SIVcpz infection of captive chimpanzees therefore shares features of both non-pathogenic and pathogenic lentivirus infections.


Ape Diseases/virology , HIV-1/physiology , Lentivirus Infections/veterinary , Lentiviruses, Primate/physiology , Pan troglodytes , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Animals , Ape Diseases/immunology , Ape Diseases/pathology , Ape Diseases/physiopathology , Autoimmune Diseases/etiology , Autoimmune Diseases/veterinary , Biomarkers/blood , CD4 Lymphocyte Count , Female , HIV-1/immunology , HIV-1/isolation & purification , Hyperplasia , Lentivirus Infections/immunology , Lentivirus Infections/physiopathology , Lentivirus Infections/virology , Lentiviruses, Primate/immunology , Lentiviruses, Primate/isolation & purification , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymph Nodes/virology , Male , Myxovirus Resistance Proteins/metabolism , Neopterin/blood , Peptide Fragments/blood , Peptide Fragments/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/blood , Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/isolation & purification , Thrombocytopenia/etiology , Thrombocytopenia/veterinary , Viral Load , beta 2-Microglobulin/blood
...