Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Comp Immunol Microbiol Infect Dis ; 111: 102202, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38852439

ABSTRACT

Avian Alpha-influenza-virus (AIV) massively affects poultry, targeting mainly the respiratory tract for virus replication. Recently, two major H5N8 and H5N1 outbreaks caused tremendous losses in Algerian poultry. The clinical symptoms that had not been seen in the past didn't prompt a rapid reaction to control the epidemics. We report here the characteristics of these outbreaks and the epidemiological status of AIV in Algeria. Following autopsy observation samples from target organs were taken and analyzed by the classical real-time reverse transcription polymerase chain reaction (RRT-PCR). Specific PCR HA and NA identification was used for subtyping H5 and N1/N8 genes. Systemic damage was observed in the upper-respiratory tracts with hemorrhagic and congestive tracheas, lungs, proventriculus, gut, and cecal tonsils were bloody. Out of 77 positive cases 13 were H5N8, 8 H5N1, and 10 H5Nx strains. These findings raise questions about the strain's pathotype considering severe organ damage and high mortality.

2.
Pol J Microbiol ; 71(4): 589-599, 2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36537059

ABSTRACT

Aflatoxin (AF)-producing fungi such as Aspergillus flavus commonly contaminate animal feeds, causing high economic losses. A. flavus is the most prevalent and produces AFB1, a potent mutagen, and carcinogen threatening human and animal health. Aspergillaceae is a large group of closely related fungi sharing number of morphological and genetic similarities that complicate the diagnosis of highly pathogenic strains. We used here morphological and molecular assays to characterize fungal isolates from animal feeds in Southwestern Algeria. These tools helped to identify 20 out of 30 Aspergillus strains, and 15 of them belonged to the Aspergillus section Flavi. Further analyses detected four out of 15 as belonging to Aspergillus flavus-parasiticus group. PCR targeting the AF genes' aflR-aflS(J) intergenic region amplified a single 674 bp amplicon in all four isolates. The amplicons were digested with a BglII endonuclease, and three specific fragments were observed for A. flavus but A. parasitucus lacked two typical fragments. Sequencing data of four amplicons confirmed the presence of the two BglII restriction sites yielding the three fragments, confirming that all four strains were A. flavus. In addition, this analysis illustrated the genetic variability within the A. flavus strains.


Subject(s)
Aflatoxins , Aspergillus flavus , Animals , Humans , Aspergillus flavus/genetics , Aspergillus , Aflatoxins/analysis , Aflatoxins/genetics , Polymerase Chain Reaction , Animal Feed
3.
Pathogens ; 11(7)2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35890043

ABSTRACT

Animal lentiviruses (LVs) have been proven to have the capacity to cross the species barrier, to adapt in the new hosts, and to increase their pathogenesis, therefore leading to the emergence of threatening diseases. However, their potential for widespread diffusion is limited by restrictive cellular factors that block viral replication in the cells of many species. In previous studies, we demonstrated that the restriction of CAEV infection of sheep choroid plexus cells was due to aberrant post-translation cleavage of the CAEV Env gp170 precursor. Later, we showed that the lack of specific receptor(s) for caprine encephalitis arthritis virus (CAEV) on the surface of human cells was the only barrier to their infection. Here, we examined whether small ruminant (SR) cells can support the replication of primate LVs. Three sheep and goat cell lines were inoculated with cell-free HIV-1 and SIVmac viral stocks or transfected with infectious molecular clone DNAs of these viruses. The two recombinant lentiviral clones contained the green fluorescent protein (GFP) reporter sequence. Infection was detected by GFP expression in target cells, and the infectious virus produced and released in the culture medium of treated cells was detected using the indicator TZM-bl cell line. Pseudotyped HIV-GFP and SIV-GFP with vesicular stomatitis virus G glycoprotein (VSV-G) allowed the cell receptors to be overcome for virus entry to further evaluate the viral replication/restriction in SR cells. As expected, neither HIV nor SIV viruses infected any of the SR cells. In contrast, the transfection of plasmid DNAs of the infectious molecular clones of both viruses in SR cells produced high titers of infectious viruses for human indicators, but not SR cell lines. Surprisingly, SR cells inoculated with HIV-GFP/VSV-G, but not SIV-GFP/VSV-G, expressed the GFP and produced a virus that efficiently infected the human indictor, but not the SR cells. Collectively, these data provide a demonstration of the lack of replication of the SIVmac genome in SR cells, while, in contrast, there was no restriction on the replication of the IV-1 genome in these cells. However, because of the lack of functional receptors to SIVmac and HIV-1 at the surface of SR cells, there is specific lentiviral entry.

4.
Vaccines (Basel) ; 10(3)2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35335093

ABSTRACT

HIV-1 remains a major public health issue worldwide in spite of efficacious antiviral therapies, but with no cure or preventive vaccine. The latter has been very challenging, as virus infection is associated with numerous escape mechanisms from host specific immunity and the correlates of protection remain incompletely understood. We have developed an innovative vaccine strategy, inspired by the efficacy of live-attenuated virus, but with the safety of a DNA vaccine, to confer both cellular and humoral responses. The CAL-SHIV-IN- lentiDNA vaccine comprises the backbone of the pathogenic SHIVKU2 genome, able to mimic the early phase of viral infection, but with a deleted integrase gene to ensure safety precluding integration within the host genome. This vaccine prototype, constitutively expressing viral antigen under the CAEV LTR promoter, elicited a variety of vaccine-specific, persistent CD4 and CD8 T cells against SIV-Gag and Nef up to 80 weeks post-immunization in cynomolgus macaques. Furthermore, these specific responses led to antiviral control of the pathogenic SIVmac251. To further improve the efficacy of this vaccine, we incorporated the IL-7 or IL-15 genes into the CAL-SHIV-IN- plasmid DNA in efforts to increase the pool of vaccine-specific memory T cells. In this study, we examined the immunogenicity of the two co-injected lentiDNA vaccines CAL-SHIV-IN- IRES IL-7 and CAL-SHIV-IN- IRES IL-15 in BALB/cJ mice and rhesus macaques and compared the immune responses with those generated by the parental vaccine CAL-SHIV-IN-. This co-immunization elicited potent vaccine-specific CD4 and CD8 T cells both in mice and rhesus macaques. Antibody-dependent cell-mediated cytotoxicity (ADCC) antibodies were detected up to 40 weeks post-immunization in both plasma and mucosal compartments of rhesus macaques and were enhanced by the cytokines.

5.
Vaccine ; 38(21): 3729-3739, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32278522

ABSTRACT

Variety of conventional vaccine strategies tested against HIV-1 have failed to induce protection against HIV acquisition or durable control of viremia. Therefore, innovative strategies that can induce long lasting protective immunity against HIV chronic infection are needed. Recently, we developed an integration-defective HIV lentiDNA vaccine that undergoes a single cycle of replication in target cells in which most viral antigens are produced. A single immunization with such lentiDNA induced long-lasting T-cell and modest antibody responses in cynomolgus macaques. Here eighteen months after this single immunization, all animals were subjected to repeated low dose intra-rectal challenges with a heterologous pathogenic SIVmac251 isolate. Although the viral set point in SIVmac-infected cynomolgus is commonly lower than that seen in Indian rhesus macaques, the vaccinated group of macaques displayed a two log reduction of peak of viremia followed by a progressive and sustained control of virus replication relative to control animals. This antiviral control correlated with antigen-specific CD4+ and CD8+ T cells with high capacity of recall responses comprising effector and central memory T cells but also memory T cell precursors. This is the first description of SIV control in NHP model infected at 18 months following a single immunization with a non-integrative single cycle lentiDNA HIV vaccine. While not delivering sterilizing immunity, our single immunization strategy with a single-cycle lentivector DNA vaccine appears to provide an interesting and safe vaccine platform that warrants further exploration.


Subject(s)
SAIDS Vaccines , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Antibodies, Viral , DNA , Immunization , Macaca mulatta , SAIDS Vaccines/genetics , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology
6.
J Vet Res ; 63(1): 27-33, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30989132

ABSTRACT

INTRODUCTION: Infection of goats with caprine arthritis encephalitis virus (CAEV) has been detected in variable proportions in many countries all over the world. Here, we investigated the seroprevalence of CAEV in goats raised in Algeria. MATERIAL AND METHODS: A serological survey was performed on serum samples from 1,313 goats, including the local breeds (Arabia and Dwarf of Kabylia) and imported European breeds (Alpine and Saanen). Blood samples were taken from goats on 38 farms distributed across four different geographical regions of Algeria. Serum samples were tested for CAEV antibodies using a commercial ELISA. RESULTS: A total of 390 serum samples were found to be positive for CAEV, giving an overall seropositivity rate of 29.7% in individual animals and 97.37% (37/38) at the goat farm level. CONCLUSION: These results provide the first large-scale serological evidence for the presence of CAEV infection in both the local and imported breeds of goats raised in Algeria, indicating that the virus infection is widespread.

7.
Virology ; 532: 69-81, 2019 06.
Article in English | MEDLINE | ID: mdl-31022666

ABSTRACT

Retroviral integrase (IN) proteins catalyze the permanent integration of the viral genome into host DNA. They can productively recruit cellular proteins, and the human Bromodomain and Extra-Terminal domain (hBET) proteins have been shown to be co-factors for integration of gamma-retroviruses such as Murine Leukemia Virus (MLV) into human cells. By using two-hybrid, co-immunoprecipitation and in vitro interaction assays, we showed that IN of the gamma- Porcine Endogenous Retrovirus-A/C (PERV IN) interacts through its C-terminal domain (CTD) with hBET proteins. We observed that PERV IN interacts with the BRD2, BRD3 and BRD4 proteins in vitro and that the BRD2 protein specifically binds and co-localizes with PERV IN protein in the nucleus of cells. We further mapped the interaction sites to the conserved Extra-Terminal (ET) domain of the hBET proteins and to several amino acids of the of the C-terminal tail of the PERV IN CTD. Finally, we determined the first experimental structure of an IN CTD - BET ET complex from small-angle X-ray scattering data (SAXS). We showed that the two factors assemble as two distinct modules linked by a short loop which confers partial flexibility. The SAXS-restrained model is structurally compatible with the binding of the PERV intasome to BRD2. Altogether, these data confirm the important role of host BET proteins in the gamma-retroviruses' targeting site and efficiency of integration.


Subject(s)
Cell Cycle Proteins/chemistry , Endogenous Retroviruses/genetics , Host-Pathogen Interactions/genetics , Integrases/chemistry , Transcription Factors/chemistry , Amino Acid Sequence , Animals , Binding Sites , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Cell Nucleus/virology , Crystallography, X-Ray , Endogenous Retroviruses/metabolism , Gene Expression , Gene Expression Regulation , HEK293 Cells , Humans , Integrases/genetics , Integrases/metabolism , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Swine , Transcription Factors/genetics , Transcription Factors/metabolism , Virus Integration
8.
Ann Biol Clin (Paris) ; 76(1): 75-80, 2018 01 01.
Article in French | MEDLINE | ID: mdl-29336321

ABSTRACT

Breast cancer is the common malignancy that affects women worldwide, but conventional risk factors account for only a small proportion of these cases. A possible viral etiology for breast cancer has been proposed and Epstein-Barr virus (EBV) is a widely studied candidate virus. The objective of this study is to determine the association of EBV infection with infiltrating ductal carcinomas (IDC). This descriptive study was carried out in the laboratory of developmental biology and differentiation, from 2012 to 2014. Of 39 cases, we determined the clinicopathological characteristics of the population. Of the 23 cases of IDC, we implemented the techniques Elisa, immunohistochemistry and in situ hybridization. To determine the serological profile, overexpression of onco-proteins EBNA-1, HER2, the mitotic index Ki67 and detection of the presence of the viral genome. The mean age is 57.40±4, SBR II predominates with 70%, pN+ (27%), RE+ (58%), RP+ (52%), HER2 (81%), Luminal A (34%), Luminal B (14%), HER2 (24%), and triple negative (28%). The serological profile of IgG VCA + in IgG EBNA-1 (87%), EBNA-1 P79 (82%) with a positive relationship between the IgG EBNA-1 and EBNA-1 P79 serology profile (p=0.001), HER2 (p=0.003) and with the molecular profile (p=0.051), EBNA-1 overexpression in (13%). The viral genome (EBER) is found in the tumors 43% representing an inverse relationship with the overexpression of Ki67 and a positive relationship with the overexpression of HER2. In our study we found an association with the presence of the EBV virus and the IDC studied.


Subject(s)
Breast Neoplasms/virology , Carcinoma, Ductal, Breast/virology , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Nuclear Antigens/isolation & purification , Herpesvirus 4, Human/isolation & purification , RNA, Viral/isolation & purification , Adult , Algeria/epidemiology , Breast Neoplasms/complications , Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/complications , Carcinoma, Ductal, Breast/epidemiology , Carcinoma, Ductal, Breast/genetics , Enzyme-Linked Immunosorbent Assay , Epstein-Barr Virus Infections/epidemiology , Epstein-Barr Virus Nuclear Antigens/analysis , Epstein-Barr Virus Nuclear Antigens/metabolism , Female , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Humans , Immunohistochemistry , In Situ Hybridization , Molecular Typing/methods , RNA, Viral/genetics , RNA, Viral/metabolism
10.
Vaccine ; 33(19): 2273-2282, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25825333

ABSTRACT

Novel HIV vaccine vectors and strategies are needed to control HIV/AIDS epidemic in humans and eradicate the infection. DNA vaccines alone failed to induce immune responses robust enough to control HIV-1. Development of lentivirus-based DNA vaccines deficient for integration and with a limited replication capacity is an innovative and promising approach. This type of vaccine mimics the early stages of virus infection/replication like the live-attenuated viruses but lacks the inconvenient integration and persistence associated with disease. We developed a novel lentivector DNA vaccine "CAL-SHIV-IN(-)" that undergoes a single round of replication in the absence of integration resulting in augmented expression of vaccine antigens in vivo. Vaccine gene expression is under control of the LTRs of a naturally attenuated lentivirus, Caprine arthritis encephalitis virus (CAEV) the natural goat lentivirus. The safety of this vaccine prototype was increased by the removal of the integrase coding sequences from the pol gene. We examined the functional properties of this lentivector DNA in cell culture and the immunogenicity in mouse models. Viral proteins were expressed in transfected cells, assembled into viral particles that were able to transduce once target permissive cells. Unlike the parental replication-competent SHIV-KU2 that was detected in DNA samples from any of the serial passage infected cells, CAL-SHIV-IN(-) DNA was detected only in target cells of the first round of infection, hence demonstrating the single cycle replication of the vaccine. A single dose DNA immunization of humanized NOD/SCID/ß2 mice showed a substantial increase of IFN-γ-ELISPOT in splenocytes compared to the former replication and integration defective Δ4SHIV-KU2 DNA vaccine.


Subject(s)
AIDS Vaccines/immunology , HIV-1/immunology , HIV-1/physiology , Vaccines, DNA/immunology , Virus Replication , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , AIDS Vaccines/isolation & purification , Animals , Enzyme-Linked Immunospot Assay , Gene Deletion , HIV Integrase/genetics , HIV-1/genetics , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Mice, Inbred BALB C , Mice, SCID , Spleen/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/isolation & purification , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/isolation & purification
11.
Vet Sci ; 2(4): 293-348, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-29061947

ABSTRACT

The emergence of human immunodeficiency virus (HIV) causing acquired immunodeficiency syndrome (AIDS) in infected humans has resulted in a global pandemic that has killed millions. HIV-1 and HIV-2 belong to the lentivirus genus of the Retroviridae family. This genus also includes viruses that infect other vertebrate animals, among them caprine arthritis-encephalitis virus (CAEV) and Maedi-Visna virus (MVV), the prototypes of a heterogeneous group of viruses known as small ruminant lentiviruses (SRLVs), affecting both goat and sheep worldwide. Despite their long host-SRLV natural history, SRLVs were never found to be responsible for immunodeficiency in contrast to primate lentiviruses. SRLVs only replicate productively in monocytes/macrophages in infected animals but not in CD4+ T cells. The focus of this review is to examine and compare the biological and pathological properties of SRLVs as prototypic Tat-independent lentiviruses with HIV-1 as prototypic Tat-dependent lentiviruses. Results from this analysis will help to improve the understanding of why and how these two prototypic lentiviruses evolved in opposite directions in term of virulence and pathogenicity. Results may also help develop new strategies based on the attenuation of SRLVs to control the highly pathogenic HIV-1 in humans.

12.
PLoS One ; 9(10): e110883, 2014.
Article in English | MEDLINE | ID: mdl-25337803

ABSTRACT

Prevention of HIV acquisition and replication requires long lasting and effective immunity. Given the state of HIV vaccine development, innovative vectors and immunization strategies are urgently needed to generate safe and efficacious HIV vaccines. Here, we developed a novel lentivirus-based DNA vector that does not integrate in the host genome and undergoes a single-cycle of replication. Viral proteins are constitutively expressed under the control of Tat-independent LTR promoter from goat lentivirus. We immunized six macaques once only with CAL-SHIV-IN- DNA using combined intramuscular and intradermal injections plus electroporation. Antigen-specific T cell responses were monitored for 47 weeks post-immunization (PI). PBMCs were assessed directly ex vivo or after 6 and 12 days of in vitro culture using antigenic and/or homeostatic proliferation. IFN-γ ELISPOT was used to measure immediate cytokine secretion from antigen specific effector cells and from memory precursors with high proliferative capacity (PHPC). The memory phenotype and functions (proliferation, cytokine expression, lytic content) of specific T cells were tested using multiparametric FACS-based assays. All immunized macaques developed lasting peripheral CD8+ and CD4+ T cell responses mainly against Gag and Nef antigens. During the primary expansion phase, immediate effector cells as well as increasing numbers of proliferating cells with limited effector functions were detected which expressed markers of effector (EM) and central (CM) memory phenotypes. These responses contracted but then reemerged later in absence of antigen boost. Strong PHPC responses comprising vaccine-specific CM and EM T cells that readily expanded and acquired immediate effector functions were detected at 40/47 weeks PI. Altogether, our study demonstrated that a single immunization with a replication-limited DNA vaccine elicited persistent vaccine-specific CM and EM CD8+ and CD4+ T cells with immediate and readily inducible effector functions, in the absence of ongoing antigen expression.


Subject(s)
AIDS Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/prevention & control , Vaccination , AIDS Vaccines/genetics , Animals , Antibodies, Viral/blood , B-Lymphocyte Subsets/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon-gamma/physiology , Lentivirus/genetics , Lentivirus/immunology , Macaca fascicularis , Male , Vaccines, DNA/genetics , Vaccines, DNA/immunology
13.
Viruses ; 5(7): 1867-84, 2013 Jul 23.
Article in English | MEDLINE | ID: mdl-23881276

ABSTRACT

Zoonotic events of simian immunodeficiency virus (SIV) from non-human primates to humans have generated the acquired immunodeficiency syndrome (AIDS), one of the most devastating infectious disease of the last century with more than 30 million people dead and about 40.3 million people currently infected worldwide. Human immunodeficiency virus (HIV-1 and HIV-2), the two major viruses that cause AIDS in humans are retroviruses of the lentivirus genus. The genus includes arthritis-encephalitis virus (CAEV) and Maedi-Visna virus (MVV), and a heterogeneous group of viruses known as small ruminant lentiviruses (SRLVs), affecting goat and sheep. Lentivirus genome integrates into the host DNA, causing persistent infection associated with a remarkable diversity during viral replication. Direct evidence of mixed infections with these two closely related SRLVs was found in both sheep and goats. The evidence of a genetic continuum with caprine and ovine field isolates demonstrates the absence of an efficient species barrier preventing cross-species transmission. In dual-infected animals, persistent infections with both CAEV and MVV have been described, and viral chimeras have been detected. This not only complicates animal trade between countries but favors the risk that highly pathogenic variants may emerge as has already been observed in the past in Iceland and, more recently, in outbreaks with virulent strains in Spain. SRLVs affecting wildlife have already been identified, demonstrating the existence of emergent viruses adapted to new hosts. Viruses adapted to wildlife ruminants may acquire novel biopathological properties which may endanger not only the new host species but also domestic ruminants and humans. SRLVs infecting sheep and goats follow a genomic evolution similar to that observed in HIV or in other lentiviruses. Lentivirus genetic diversity and host factors leading to the establishment of naturally occurring virulent versus avirulent infections, in addition to the emergence of new strains, challenge every aspect of SRLV control measures for providing efficient tools to prevent the transmission of diseases between wild ungulates and livestock.


Subject(s)
Host Specificity , Lentiviruses, Ovine-Caprine/physiology , Adaptation, Biological , Animals , Arthritis-Encephalitis Virus, Caprine/genetics , Disease Outbreaks , Goats , Iceland/epidemiology , Lentivirus Infections/veterinary , Lentivirus Infections/virology , Lentiviruses, Ovine-Caprine/genetics , Recombination, Genetic , Sheep , Spain/epidemiology , Visna-maedi virus/genetics
14.
Comp Immunol Microbiol Infect Dis ; 36(4): 397-404, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23623734

ABSTRACT

The transmission of CAEV from male goats has not been well studied and the target cells that support viral replication are not well characterized. Epididymal epithelial cells (EECs) are important and play a key role in the fertility and motility of spermatozoa. During their transit, spermatozoa incorporate several EEC-produced proteins into their plasma membranes to stabilize them and prevent premature acrosomal reaction. This intimate interaction between spermatozoa and EECs may increase the likelihood of the infection of semen with CAEV if epididymal tissue is productively infected and sheds the virus into the duct. The aim of this study was to examine whether goat EECs are susceptible to CAEV infection in tissue culture. Cells were isolated from epididymides obtained from goats that were sampled from a certified-CAEV-free herd. Cultured cells were then inoculated with a molecularly-cloned isolate of CAEV (CAEV-pBSCA). Inoculated cells developed cytopathic effects (CPE), showing numerous multinucleated giant cells (MGC) in cell-culture monolayers. Expression of CAEV proteins was detected by immunofluorescence using an anti-p28, Gag-specific antibody. The culture medium of inoculated cells was shown to contain high titers (10(6) tissue culture infectious doses 50 per ml (TCID50/ml)) of infectious, cytopathic virus when assayed using indicator goat synovial membrane (GSM) cells. Our findings clearly demonstrate that cells of the buck genital tract are targets of CAEV and are thus a potential reservoir that sheds infectious CAEV into the semen of infected animals. These data suggest the use of sperm from CAEV-free goat males for artificial insemination in genetic selection programs to minimize CAEV dissemination.


Subject(s)
Arthritis-Encephalitis Virus, Caprine/immunology , Epididymis/virology , Goat Diseases/virology , Lentivirus Infections/veterinary , Animals , Arthritis-Encephalitis Virus, Caprine/genetics , Cytopathogenic Effect, Viral/immunology , DNA, Viral/chemistry , DNA, Viral/genetics , Epididymis/cytology , Epididymis/immunology , Epithelial Cells , Fluorescent Antibody Technique , Goat Diseases/immunology , Goats , Lentivirus Infections/immunology , Lentivirus Infections/virology , Male , Polymerase Chain Reaction/veterinary , Virus Replication/immunology
15.
Vaccine ; 30(19): 2956-62, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22387218

ABSTRACT

Increasing the safety and the efficacy of existing HIV vaccines is one of the strategies that could help to promote the development of a vaccine for human use. We developed a HIV DNA vaccine (Δ4-SHIVKU2) that has been shown to induce potent polyfunctional HIV-specific T cell responses following a single dose immunization of mice and macaques. Δ4-SHIVKU2 also induced protection when immunized macaques were challenged with homologous pathogenic viruses. In the present study, our aim was to examine whether a chimeric HIV DNA vaccine (CAL-Δ4-SHIVKU2) whose genome is driven by the LTR of the goat lentivirus, caprine arthritis encephalitis (CAEV) expresses efficiently the vaccine antigens and induces potent immune responses in animal models for HIV vaccine. Data of radioimmunoprecipitation assays clearly show that this chimeric genome drives efficient expression of all HIV antigens in the construct. In addition, evaluation of the p24 Gag protein in the supernatant of HEK-293-T cells transfected in parallel with Δ4-SHIVKU2 and CAL-Δ4-SHIVKU2 showed no difference suggesting that these two LTRs are inducing equally the expression of the viral genes. Immunization of mice and macaques using our single dose immunization regimen resulted in induction of similar IFN-γ ELISPOT responses in Δ4-SHIVKU2- and CAL-Δ4-SHIVKU2-treated mice. Similar profiles of T cell responses were also detected both in mice and macaques when multiparametric flow cytometry analyses were performed. Since CAEV LTR is not dependent of Tat to drive viral gene expression and is not functional for integration with HIV integrase, this new vector increases the safety and efficacy of our vaccine vectors and vaccination strategy.


Subject(s)
AIDS Vaccines/immunology , Arthritis-Encephalitis Virus, Caprine/genetics , HIV/immunology , Terminal Repeat Sequences , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/genetics , Animals , Cell Line , Enzyme-Linked Immunospot Assay , Female , Gene Expression Profiling , HIV/genetics , HIV Core Protein p24/biosynthesis , Humans , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Macaca mulatta , Mice , Mice, Inbred BALB C , Vaccines, DNA/administration & dosage
16.
Vet Res ; 43: 5, 2012 Jan 25.
Article in English | MEDLINE | ID: mdl-22276529

ABSTRACT

The aim of this study was to determine, using immunofluorescence and in situ hybridization, whether CAEV is capable of infecting goat uterine epithelial cells in vivo. Five CAEV seropositive goats confirmed as infected using double nested polymerase chain reaction (dnPCR) on leucocytes and on vaginal secretions were used as CAEV positive goats. Five CAEV-free goats were used as controls. Samples from the uterine horn were prepared for dnPCR, in situ hybridization, and immunofluorescence. The results from dnPCR confirmed the presence of CAEV proviral DNA in the uterine horn samples of infected goats whereas no CAEV proviral DNA was detected in samples taken from the uninfected control goats. The in situ hybridization probe was complementary to part of the CAEV gag gene and confirmed the presence of CAEV nucleic acids in uterine samples. The positively staining cells were seen concentrated in the mucosa of the lamina propria of uterine sections. Finally, laser confocal analysis of double p28/cytokeratin immunolabelled transverse sections of CAEV infected goat uterus, demonstrated that the virus was localized in glandular and epithelial cells. This study clearly demonstrates that goat uterine epithelial cells are susceptible to CAEV infection in vivo. This finding could help to further our understanding of the epidemiology of CAEV, and in particular the possibility of vertical transmission.


Subject(s)
Arthritis-Encephalitis Virus, Caprine/isolation & purification , Epithelial Cells/virology , Goat Diseases/virology , Lentivirus Infections/veterinary , Proviruses/isolation & purification , Uterine Diseases/veterinary , Uterus/virology , Animals , DNA, Viral/blood , DNA, Viral/metabolism , Female , Fluorescent Antibody Technique/veterinary , Goats , In Situ Hybridization/veterinary , Lentivirus Infections/virology , Microscopy, Confocal/veterinary , Polymerase Chain Reaction/veterinary , Uterine Diseases/virology
17.
J Virol ; 84(3): 1243-53, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923181

ABSTRACT

The optimization of immune responses (IR) induced by HIV DNA vaccines in humans is one of the great challenges in the development of an effective vaccine against AIDS. Ideally, this vaccine should be delivered in a single dose to immunize humans. We recently demonstrated that the immunization of mice with a single dose of a DNA vaccine derived from pathogenic SHIV(KU2) (Delta4SHIV(KU2)) induced long-lasting, potent, and polyfunctional HIV-specific CD8(+) T-cell responses (G. Arrode, R. Hegde, A. Mani, Y. Jin, Y. Chebloune, and O. Narayan, J. Immunol. 178:2318-2327, 2007). In the present work, we expanded the characterization of the IR induced by this DNA immunization protocol to rhesus macaques. Animals immunized with a single high dose of Delta4SHIV(KU2) DNA vaccine were monitored longitudinally for vaccine-induced IR using multiparametric flow cytometry-based assays. Interestingly, all five immunized macaques developed broad and polyfunctional HIV-specific T-cell IR that persisted for months, with an unusual reemergence in the blood following an initial decline but in the absence of antibody responses. The majority of vaccine-specific CD4(+) and CD8(+) T cells lacked gamma interferon production but showed high antigen-specific proliferation capacities. Proliferative CD8(+) T cells expressed the lytic molecule granzyme B. No integrated viral vector could be detected in mononuclear cells from immunized animals, and this high dose of DNA did not induce any detectable autoimmune responses against DNA. Taken together, our comprehensive analysis demonstrated for the first time the capacity of a single high dose of HIV DNA vaccine alone to induce long-lasting and polyfunctional T-cell responses in the nonhuman primate model, bringing new insights for the design of future HIV vaccines.


Subject(s)
AIDS Vaccines/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Vaccines, DNA/administration & dosage , AIDS Vaccines/immunology , Animals , B-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interferon-gamma/biosynthesis , Macaca mulatta , Mice , Plasmids , Vaccines, DNA/immunology
18.
Virology ; 383(1): 93-102, 2009 Jan 05.
Article in English | MEDLINE | ID: mdl-19007964

ABSTRACT

The small ruminant lentiviruses, caprine arthritis-encephalitis virus (CAEV) and maedi visna virus (MVV) naturally cause inflammatory disease in goats and sheep, provoking chronic lesions in several different organs. We have previously demonstrated that in vitro infection of caprine cells by CAEV induces apoptosis through the intrinsic pathway (Rea-Boutrois, A., Pontini, G., Greenland, T., Mehlen, P., Chebloune, Y., Verdier, G. and Legras-Lachuer, C. 2008). In the present study, we used Tat deleted viruses and SLRV Tat-expression vectors to show that the SRLV Tat proteins are responsible for this apoptosis. We have also studied the activation of caspases-3, -8 and -9 by fluorescent assays in caprine cells expressing SRLV Tat proteins, and the effects of transfected dominant negative variants of these caspases, to show that Tat-associated apoptosis depends on activation of caspases-3 and -9, but not -8. A simultaneous disruption of mitochondrial membrane potential indicates an involvement of the mitochondrial pathway.


Subject(s)
Apoptosis , Arthritis-Encephalitis Virus, Caprine/pathogenicity , Gene Products, tat/toxicity , Animals , Arthritis-Encephalitis Virus, Caprine/genetics , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line , Gene Deletion , Gene Products, tat/genetics , Goats , Membrane Potential, Mitochondrial/physiology
19.
Glia ; 57(7): 734-43, 2009 May.
Article in English | MEDLINE | ID: mdl-18985732

ABSTRACT

HIV encephalitis (HIVE), the pathologic correlate of HIV-associated dementia (HAD) is characterized by astrogliosis, cytokine/chemokine dysregulation, and neuronal degeneration. Increasing evidence suggests that inflammation is actively involved in the pathogenesis of HAD. In fact, the severity of HAD/HIVE correlates more closely with the presence of activated glial cells than with the presence and amount of HIV-infected cells in the brain. Astrocytes, the most numerous cell type within the brain, provide an important reservoir for the generation of inflammatory mediators, including interferon-gamma inducible peptide-10 (CXCL10), a neurotoxin and a chemoattractant, implicated in the pathophysiology of HAD. Additionally, the proinflammatory cytokines, IFN-gamma and TNF-alpha, are also markedly increased in CNS tissues during HIV-1 infection. In this study, we hypothesized that the interplay of host cytokines and HIV-1 could lead to enhanced expression of the toxic chemokine, CXCL10. Our findings demonstrate a synergistic induction of CXCL10 mRNA and protein in human astrocytes exposed to HIV-1 and the proinflammatory cytokines. Signaling molecules, including JAK, STATs, MAPK (via activation of Erk1/2, AKT, and p38), and NF-kappaB were identified as instrumental in the synergistic induction of CXCL10. Understanding the mechanisms involved in HIV-1 and cytokine-mediated up-regulation of CXCL10 could aid in the development of therapeutic modalities for HAD.


Subject(s)
Astrocytes/metabolism , Chemokine CXCL10/metabolism , Cytokines/metabolism , HIV-1/physiology , Astrocytes/immunology , Astrocytes/virology , Blotting, Western , Cell Line , Chemokine CXCL10/genetics , Enzyme-Linked Immunosorbent Assay , Gene Expression , Humans , Immunohistochemistry , Interferon-gamma/metabolism , NF-kappa B/metabolism , Polymerase Chain Reaction , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
20.
Virology ; 383(1): 103-11, 2009 Jan 05.
Article in English | MEDLINE | ID: mdl-18986665

ABSTRACT

Live-attenuated viruses derived from SIV and SHIV have provided the most consistent protection against challenge with pathogenic viruses, but concerns regarding their long-term safety and efficacy have hampered their clinical usefulness. We report a longitudinal study in which we evaluated the long-term safety and efficacy of DeltavpuSHIV(PPC), a live virus vaccine derived from SHIV(PPC). Macaques were administered two inoculations of DeltavpuSHIV(PPC), three years apart, and followed for eight years. None of the five vaccinated macaques developed an AIDS-like disease from the vaccine. At eight years, macaques were challenged with pathogenic SIV and SHIV. None of the four macaques with detectable cellular-mediated immunity prior to challenge had detectable viral RNA in the plasma. This study demonstrates that multiple inoculations of a live vaccine virus can be used safely and can significantly extend the efficacy of the vaccine, as compared to a single inoculation, which is efficacious for approximately three years.


Subject(s)
SAIDS Vaccines/adverse effects , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Animals , Immunization, Secondary , Leukocytes, Mononuclear/immunology , Longitudinal Studies , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/prevention & control , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Viremia/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...