Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 428
Filter
1.
BMC Vet Res ; 20(1): 195, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741095

ABSTRACT

Small ruminant lentiviruses (SRLVs) are widespread and infect goats and sheep. Several reports also suggest that SRLVs can infect wild ruminants. The presence of specific antibodies against SRLVs has been identified in wild ruminants from Poland, but no studies have been conducted to detect proviral DNA of SRLVs in these animals. Therefore, the purpose of this study was to examine samples from Polish wild ruminants to determine whether these animals can serve as reservoirs of SRLVs under natural conditions. A total of 314 samples were tested from red deer (n = 255), roe deer (n = 52) and fallow deer (n = 7) using nested real-time PCR. DNA from positive real-time PCR samples was subsequently used to amplify a CA fragment (625 bp) of the gag gene, a 1.2 kb fragment of the pol gene and an LTR-gag fragment. Three samples (0.95%) were positive according to nested real-time PCR using primers and probe specific for CAEV (SRLV group B). All the samples were negative for the primers and probe specific for MVV (SRLV A group). Only SRLV LTR-gag sequences were obtained from two red deer. Phylogenetic analysis revealed that these sequences were more closely related to CAEV than to MVV. Our results revealed that deer can carry SRLV proviral sequences and therefore may play a role in the epidemiology of SRLVs. To our knowledge, this is the first study describing SRLV sequences from red deer.


Subject(s)
DNA, Viral , Deer , Lentivirus Infections , Proviruses , Animals , Deer/virology , Poland/epidemiology , Proviruses/genetics , Lentivirus Infections/veterinary , Lentivirus Infections/virology , Lentivirus Infections/epidemiology , DNA, Viral/genetics , Lentivirus/isolation & purification , Lentivirus/genetics , Lentivirus/classification , Phylogeny , Real-Time Polymerase Chain Reaction/veterinary
2.
Viruses ; 16(4)2024 Mar 31.
Article in English | MEDLINE | ID: mdl-38675890

ABSTRACT

The high genetic heterogeneity of small ruminant lentiviruses (SRLV) renders the genetic characterization of the circulating strains crucial for the epidemiological investigation and the designation of effective diagnostic tools. In Greece, research data regarding the genetic diversity of the circulating SRLV strains is scarce, hindering the implementation of efficient surveillance and control programs. The objective of the study was to genetically characterize SRLV strains isolated from intensive dairy sheep farms in Greece and evaluate the variability of the immunodominant regions of the capsid protein. For this reason, a total of 12 SRLV-infected animals from four intensive dairy sheep farms with purebred Chios and Lacaune ewes were used for the amplification and sequencing of an 800 bp gag-pol fragment. The phylogenetic analyses revealed a breed-related circulation of strains; Chios ewes were infected with strains belonging exclusively to a separate group of genotype A, whereas strains belonging to subtype B2 were isolated from Lacaune ewes. Immunodominant epitopes of capsid protein were quite conserved among the strains of the same genotype, except for the Major Homology Region which showed some unique mutations with potential effects on viral evolution. The present study contributes to the extension of the current knowledge regarding the genetic diversity of SRLV strains circulating in sheep in Greece. However, broader genetic characterization studies are warranted for the exploration of possible recombinant events and the more comprehensive classification of the circulating strains.


Subject(s)
Genetic Variation , Genotype , Lentivirus Infections , Phylogeny , Sheep Diseases , Animals , Sheep , Greece , Sheep Diseases/virology , Lentivirus Infections/veterinary , Lentivirus Infections/virology , Female , Capsid Proteins/genetics , Lentivirus/genetics , Lentivirus/isolation & purification , Lentivirus/classification
3.
Eur J Med Res ; 27(1): 5, 2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35022077

ABSTRACT

BACKGROUND: Intervertebral disc degeneration (IDD) is a natural progression of age-related processes. Associated with IDD, degenerative disc disease (DDD) is a pathologic condition implicated as a major cause of chronic lower back pain, which can have a severe impact on the quality of life of patients. As degeneration progression is associated with elevated levels of inflammatory cytokines, enhanced aggrecan and collagen degradation, and changes in the disc cell phenotype. The purpose of this study was to investigate the biological and cytological characteristics of rabbit nucleus pulposus mesenchymal stem cells (NPMSCs)-a key factor in IDD-and to determine the effect of the growth and differentiation factor-5 (GDF5) on the differentiation of rabbit NPMSCs transduced with a lentivirus vector. METHODS: An in vitro culture model of rabbit NPMSCs was established and NPMSCs were identified by flow cytometry (FCM) and quantitative real-time PCR (qRT-PCR). Subsequently, NPMSCs were randomly divided into three groups: a transfection group (the lentiviral vector carrying GDF5 gene used to transfect NPMSCs); a control virus group (the NPMSCs transfected with an ordinary lentiviral vector); and a normal group (the NPMSCs alone). FCM, qRT-PCR, and western blot (WB) were used to detect the changes in NPMSCs. RESULTS: The GDF5-transfected NPMSCs displayed an elongated shape, with decreased cell density, and significantly increased GDF5 positivity rate in the transfected group compared to the other two groups (P < 0.01). The mRNA levels of Krt8, Krt18, and Krt19 in the transfected group were significantly higher in comparison with the other two groups (P < 0.01), and the WB results were consistent with that of qRT-PCR. CONCLUSIONS: GDF5 could induce the differentiation of NPMSCs. The lentiviral vector carrying the GDF5 gene could be integrated into the chromosome genome of NPMSCs and promoted differentiation of NPMSCs into nucleus pulposus cells. Our findings advance the development of feasible and effective therapies for IDD.


Subject(s)
Gene Expression Regulation , Growth Differentiation Factor 5/genetics , Lentivirus Infections/virology , Lentivirus , Mesenchymal Stem Cells/cytology , Nucleus Pulposus/metabolism , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Growth Differentiation Factor 5/biosynthesis , Lentivirus Infections/metabolism , Lentivirus Infections/pathology , Mesenchymal Stem Cells/virology , Nucleus Pulposus/pathology , Nucleus Pulposus/virology , Rabbits
4.
Viruses ; 13(12)2021 11 23.
Article in English | MEDLINE | ID: mdl-34960606

ABSTRACT

Small ruminant lentiviruses (SRLV) are viruses that retro-transcribe RNA to DNA and show high rates of genetic variability. SRLV affect animals with strains specific for each host species (sheep or goats), resulting in a series of clinical manifestations depending on the virulence of the strain, the host's genetic background and farm production system. The aim of this work was to present an up-to-date overview of the genomic epidemiology and genetic diversity of SRLV in Italy over time (1998-2019). In this study, we investigated 219 SRLV samples collected from 17 different Italian regions in 178 geographically distinct herds by CEREL. Our genetic study was based on partial sequencing of the gag-pol gene (800 bp) and phylogenetic analysis. We identified new subtypes with high heterogeneity, new clusters and recombinant forms. The genetic diversity of Italian SRLV strains may have diagnostic and immunological implications that affect the performance of diagnostic tools. Therefore, it is extremely important to increase the control of genomic variants to improve the control measures.


Subject(s)
Lentivirus Infections , Lentivirus/classification , Ruminants/virology , Animals , Goat Diseases/virology , Goats , Italy/epidemiology , Lentivirus Infections/epidemiology , Lentivirus Infections/veterinary , Lentivirus Infections/virology , Sheep , Sheep Diseases/virology
5.
Viruses ; 13(12)2021 12 16.
Article in English | MEDLINE | ID: mdl-34960798

ABSTRACT

Small ruminant lentiviruses (SRLVs) are a group of highly divergent viruses responsible for global infection in sheep and goats. In a previous study we showed that SRLV strains found in mixed flocks in Poland belonged to subtype A13 and A18, but this study was restricted only to the few flocks from Malopolska region. The present work aimed at extending earlier findings with the analysis of SRLVs in mixed flocks including larger numbers of animals and flocks from different part of Poland. On the basis of gag and env sequences, Polish SRLVs were assigned to the subtypes B2, A5, A12, and A17. Furthermore, the existence of a new subtypes, tentatively designed as A23 and A24, were described for the first time. Subtypes A5 and A17 were only found in goats, subtype A24 has been detected only in sheep while subtypes A12, A23, and B2 have been found in both sheep and goats. Co-infection with strains belonging to different subtypes was evidenced in three sheep and two goats originating from two flocks. Furthermore, three putative recombination events were identified within gag and env SRLVs sequences derived from three sheep. Amino acid (aa) sequences of immunodominant epitopes in CA protein were well conserved while Major Homology Region (MHR) had more alteration showing unique mutations in sequences of subtypes A5 and A17. In contrast, aa sequences of surface glycoprotein exhibited higher variability confirming type-specific variation in the SU5 epitope. The number of potential N-linked glycosylation sites (PNGS) ranged from 3 to 6 in respective sequences and were located in different positions. The analysis of LTR sequences revealed that sequences corresponding to the TATA box, AP-4, AML-vis, and polyadenylation signal (poly A) were quite conserved, while considerable alteration was observed in AP-1 sites. Interestingly, our results revealed that all sequences belonging to subtype A17 had unique substitution T to A in the fifth position of TATA box and did not have a 11 nt deletion in the R region which was noted in other sequences from Poland. These data revealed a complex picture of SRLVs population with ovine and caprine strains belonging to group A and B. We present strong and multiple evidence of dually infected sheep and goats in mixed flocks and present evidence that these viruses can recombine in vivo.


Subject(s)
Goats/virology , Lentivirus Infections/transmission , Lentivirus/genetics , Recombination, Genetic , Sheep/virology , Animals , Lentivirus/classification , Lentivirus Infections/virology , Phylogeny , Terminal Repeat Sequences
6.
Bioengineered ; 12(2): 12498-12508, 2021 12.
Article in English | MEDLINE | ID: mdl-34927536

ABSTRACT

At present, the mechanism of siSCN9A in Vincristine (VCR)-induced neuropathic pain (NP) is still unclear. This study aimed to explore the analgesic mechanism of lentivirus-siSCN9A (LV-siSCN9A) infected neurons against NP. 40 male Sprague-Dawley (SD) rats were divided into a control group (injected with normal saline), a model group (VCR-induced NP model), a LV-SC group (NP model mice were injected with LV-SC-infected dorsal root ganglia (DRG) neuron cells under the microscope), and a LV-siSCN9A group (NP model mice were injected with LV-siSCN9A-infected DRG neuron cells under the microscope, with 10 rats in each group. The changes of mechanical withdrawal threshold (MWT) and thermal withdrawal latency (TWL) of rats in different groups were detected by behavior testing, the Nav1.7 changes in each group were detected by immunofluorescence double standard and Western-blot method. It was found that compared with the control group, the MWT and TWL of the rats in model group were significantly decreased (P < 0.05), and the expression levels of Nav1.7 messenger ribonucleic acid (mRNA) and proteins were significantly increased (P < 0.05). Compared with LV-SC group, the MWT and TWL of rats in LV-siSCN9A group were significantly increased (P < 0.05), the expression levels of Nav1.7 mRNA and proteins were significantly decreased (P < 0.05), and the CGRP expression of spinal dorsal horn was significantly decreased. It was concluded that the LV-siSCN9A infected neurons could play an analgesic role by down-regulating Nav1.7 expression induced by VCR in NP model.


Subject(s)
Lentivirus Infections/virology , Lentivirus/pathogenicity , Neuralgia/chemically induced , Neuralgia/virology , Neurons/drug effects , Neurons/virology , Vincristine/pharmacology , Analgesia/methods , Animals , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/virology , Lentivirus Infections/genetics , Male , Mice , NAV1.7 Voltage-Gated Sodium Channel/genetics , Neuralgia/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
7.
Sci Rep ; 11(1): 16201, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376765

ABSTRACT

Optical spectroscopic techniques have been commonly used to detect the presence of biofilm-forming pathogens (bacteria and fungi) in the agro-food industry. Recently, near-infrared (NIR) spectroscopy revealed that it is also possible to detect the presence of viruses in animal and vegetal tissues. Here we report a platform based on visible and NIR (VNIR) hyperspectral imaging for non-contact, reagent free detection and quantification of laboratory-engineered viral particles in fluid samples (liquid droplets and dry residue) using both partial least square-discriminant analysis and artificial feed-forward neural networks. The detection was successfully achieved in preparations of phosphate buffered solution and artificial saliva, with an equivalent pixel volume of 4 nL and lowest concentration of 800 TU·[Formula: see text]L-1. This method constitutes an innovative approach that could be potentially used at point of care for rapid mass screening of viral infectious diseases and monitoring of the SARS-CoV-2 pandemic.


Subject(s)
Image Processing, Computer-Assisted/methods , Lentivirus Infections/diagnosis , Molecular Diagnostic Techniques/methods , Spectroscopy, Near-Infrared/methods , HEK293 Cells , Humans , Image Processing, Computer-Assisted/standards , Lentivirus/isolation & purification , Lentivirus/pathogenicity , Lentivirus Infections/virology , Molecular Diagnostic Techniques/standards , Point-of-Care Systems , Saliva/virology , Sensitivity and Specificity , Spectroscopy, Near-Infrared/standards
8.
Pol J Vet Sci ; 24(2): 313-317, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34250784

ABSTRACT

Small ruminant lentivirus (SRLV), which causes caprine arthritis encephalitis in goats and ovine progressive pneumonia (maedi-visna disease) in sheep, is classified in genus Lentiviruses belonging to Retroviridae family. It persists in infected goats and sheep, which mostly are sub- clinical. A serological survey was conducted to determine the prevalence of small ruminant lentivirus infection in Thai goat population. Serum samples were taken from 1,925 goats distributed throughout the country, then they were tested for the presence of SRLV antibodies using commercial indirect enzyme-linked immunosorbent assay (ELISA) test kits. Results revealed that a total of 68 goats were found seropositive, representing the apparent prevalence and true prevalence of 3.57% and 2.60%, respectively. The seroprevalence, revealed in this study, was lower than in the previous reports. The decreasing of seroprevalence might be caused by successful control strategies from Department of Livestock Development (DLD).


Subject(s)
Goat Diseases/virology , Lentivirus Infections/veterinary , Lentiviruses, Ovine-Caprine/isolation & purification , Animals , Goat Diseases/epidemiology , Goats , Lentivirus Infections/epidemiology , Lentivirus Infections/virology , Seroepidemiologic Studies , Thailand/epidemiology
9.
Viruses ; 13(6)2021 06 07.
Article in English | MEDLINE | ID: mdl-34200141

ABSTRACT

The transmission of viruses from animal hosts into humans have led to the emergence of several diseases. Usually these cross-species transmissions are blocked by host restriction factors, which are proteins that can block virus replication at a specific step. In the natural virus host, the restriction factor activity is usually suppressed by a viral antagonist protein, but this is not the case for restriction factors from an unnatural host. However, due to ongoing viral evolution, sometimes the viral antagonist can evolve to suppress restriction factors in a new host, enabling cross-species transmission. Here we examine the classical case of this paradigm by reviewing research on APOBEC3 restriction factors and how they can suppress human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV). APOBEC3 enzymes are single-stranded DNA cytidine deaminases that can induce mutagenesis of proviral DNA by catalyzing the conversion of cytidine to promutagenic uridine on single-stranded viral (-)DNA if they escape the HIV/SIV antagonist protein, Vif. APOBEC3 degradation is induced by Vif through the proteasome pathway. SIV has been transmitted between Old World Monkeys and to hominids. Here we examine the adaptations that enabled such events and the ongoing impact of the APOBEC3-Vif interface on HIV in humans.


Subject(s)
APOBEC Deaminases/genetics , Host-Pathogen Interactions/genetics , Lentivirus Infections/genetics , Lentivirus Infections/transmission , Lentiviruses, Primate/physiology , Viral Zoonoses/transmission , Animals , Gene Products, vif/chemistry , Gene Products, vif/metabolism , HIV Infections/genetics , HIV Infections/transmission , HIV Infections/virology , HIV-1/physiology , Humans , Lentivirus Infections/virology , Protein Binding , Protein Isoforms , Structure-Activity Relationship , vif Gene Products, Human Immunodeficiency Virus/chemistry , vif Gene Products, Human Immunodeficiency Virus/metabolism
10.
Vet Clin North Am Food Anim Pract ; 37(1): 199-208, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33541699

ABSTRACT

Small ruminant lentiviruses (SLRVs) have been recognized throughout the world for decades. SLRVs are a heterogenous group of viruses that can infect sheep, goats, and wild ruminants. Evidence supports cross-species infection. These viruses cause lifelong infections where they target specific organs, which can result in production losses due to diminished milk production, consequential increases in neonatal death and diminished growth, and premature culling of prime age animals. No vaccine or treatments have proved effective. Control programs rely on an understanding of viral transmission and application of highly sensitive, specific, and frequent testing regimens.


Subject(s)
Goat Diseases/virology , Lentivirus Infections/veterinary , Lentiviruses, Ovine-Caprine/physiology , Sheep Diseases/virology , Animals , Goats , Lentivirus Infections/virology , Lentiviruses, Ovine-Caprine/pathogenicity , Ruminants , Sheep
11.
Vet Microbiol ; 252: 108931, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33271419

ABSTRACT

Despite SRLV infection being endemic in Mexico, there is little information regarding which genotypes are present. We compared serotyping and PCR-sequencing results from sheep and goats infected with SRLV. We separated plasma and peripheral blood leukocytes (PBL) from 1940 blood samples from sheep and goats from 12 states across Mexico. To detect SRLV infection, we tested plasma samples using two commercial ELISA kits (VMRD and Eradikit SRLV Screening). Then, we serotyped the infecting virus (A/ B) using Eradikit SRLV Genotyping. PBL DNA was used to detect the proviral genome via PCR. Positive amplicons were sequenced to identify viral genotypes using a phylogenetic analysis. Also, we analysed for residues differences in the sequences of a capsid epitope between genotypes. The serological results indicated a higher detection of seropositive animals using the VMRD ELISA compared to Eradikit, with 21 % and 15.3 % more in sheep and goats respectively. Only 25.7 % of the ELISA serotyping results matched those from PCR-sequencing. PCR-sequencing was able to identify genotype A, B and coinfections in animals classified as indeterminate by the ELISA test. This lack of sensitivity may be related to the lack of epitopes from the matrix and transmembrane peptides used by ELISA screening. Sequences analysis revealed that SRLVs found in sheep cluster with genetic subtypes A2 and B1, while those in goats cluster with subtypes A1 and B1. Serotyping did not prove to be an adequate method for predicting the viral genotype (A and / or B) in infections caused by SRLV.


Subject(s)
Antibodies, Viral/blood , Goat Diseases/virology , Lentivirus Infections/veterinary , Lentivirus/immunology , Sheep Diseases/virology , Animals , Enzyme-Linked Immunosorbent Assay/veterinary , Genotype , Goats , Lentivirus/genetics , Lentivirus/isolation & purification , Lentivirus Infections/virology , Phylogeny , Polymerase Chain Reaction/veterinary , Ruminants , Sensitivity and Specificity , Serotyping/veterinary , Sheep
12.
J Virol ; 95(2)2020 12 22.
Article in English | MEDLINE | ID: mdl-33115880

ABSTRACT

Aluminum (Al)-based salts are widely used adjuvants in ruminants and other species to strengthen the immune response elicited against vaccine antigen(s). However, they can lead to the formation of long-lasting granulomas composed of abundant activated macrophages. Small ruminant lentiviruses (SRLV) are widely distributed macrophage-tropic retroviruses that cause persistent infections in sheep and goats. Infected monocytes/macrophages and dendritic cells establish an inflammatory microenvironment that eventually leads to clinical manifestations. The aim of this work was to study the effect of Al-induced granulomas in the replication and pathogenesis of SRLV. Eleven adult, naturally SRLV-infected sheep showing clinical arthritis were distributed in vaccine (n = 6), adjuvant-only (n = 3), and control (n = 2) groups and inoculated with commercial Al-based vaccines, Al hydroxide adjuvant alone, or phosphate-buffered saline, respectively. In vitro studies demonstrated viral replication in Al-induced granulomas in 5 out of 10 sheep. Immunohistochemistry (IHC) evinced granular, intracytoplasmic SRLV presence in macrophages within granulomas. Viral sequences obtained from granulomas, blood monocytes, and other tissues were highly similar in most animals, suggesting virus circulation among body compartments. However, notable differences between isolated strains in granulomas and other tissues in specific animals were also noted. Interestingly, the B2 subtype was the most commonly found SRLV genotype, reaching a wider body distribution than previously described. Recombination events between genotypes B2 and A3 along the gag region were identified in two sheep. Our results indicate that Al-hydroxide-derived granulomas may represent an ideal compartment for SRLV replication, perhaps altering natural SRLV infection by providing a new, suitable target tissue.IMPORTANCE Granulomas are inflammation-derived structures elicited by foreign bodies or certain infections. Aluminum adjuvants included in vaccines induce granulomas in many species. In sheep, these are persistent and consist of activated macrophages. Small ruminant lentiviruses (SRLV), which are macrophage-tropic lentiviruses, cause a chronic wasting disease affecting animal welfare and production. Here, we studied the occurrence of SRLV in postvaccination granulomas retrieved from naturally infected ewes after vaccination or inoculation with aluminum only. SRLV infection was confirmed in granulomas by identification of viral proteins, genomic fragments, and enzymatic activity. The infecting SRLV strain, previously found exclusively in carpal joints, reached the central nervous system, suggesting that occurrence of SRLV in postvaccination granulomas may broaden tissue tropism. SRLV recombination was detected in inoculated animals, a rare event in sheep lentiviruses. Potentially, virus-host interactions within granulomas may modify viral pathogenesis and lead to more widespread infection.


Subject(s)
Adjuvants, Immunologic/adverse effects , Aluminum Hydroxide/adverse effects , Arthritis-Encephalitis Virus, Caprine/physiology , Granuloma/veterinary , Lentivirus Infections/veterinary , Sheep Diseases/virology , Virus Replication/drug effects , Animals , Arthritis-Encephalitis Virus, Caprine/classification , Arthritis-Encephalitis Virus, Caprine/drug effects , Arthritis-Encephalitis Virus, Caprine/isolation & purification , Genotype , Granuloma/chemically induced , Granuloma/virology , Lentivirus Infections/virology , Macrophages/drug effects , Macrophages/virology , Phylogeny , Recombination, Genetic , Sheep , Sheep Diseases/chemically induced , Viral Tropism
13.
EBioMedicine ; 60: 103008, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32979832

ABSTRACT

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.


Subject(s)
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
14.
Sci Rep ; 10(1): 13740, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792615

ABSTRACT

Lentiviral modification of hematopoietic stem cells (HSCs) paved the way for in vivo experimentation and therapeutic approaches in patients with genetic disease. A disadvantage of this method is the use of a ubiquitous promoter leads not only to genetic modification of the leukocyte subset of interest e.g. T-cells, but also all other subsequent leukocyte progeny of the parent HSCs. To overcome this limitation we tested a bicistronic lentivirus, enabling subset specific modifications. Designed novel lentiviral constructs harbor a global promoter (mPGK) regulating mCherry for HSCs selection and a T-cell specific promoter upstream of eGFP. Two T-cell specific promoters were assessed: the distal Lck-(dLck) and the CD3δ-promoter. Transduced HSCs were FACS sorted by mCherry expression and transferred into sublethally irradiated C57/BL6 mice. Successful transplantation and T-cell specific expression of eGFP was monitored by peripheral blood assessment. Furthermore, recruitment response of lentiviral engineered leukocytes to the site of inflammation was tested in a peritonitis model without functional impairment. Our constructed lentivirus enables fast generation of subset specific leukocyte transgenesis as shown in T-cells in vivo and opens new opportunities to modify other HSCs derived subsets in the future.


Subject(s)
Hematopoietic Stem Cells/virology , Lentivirus Infections/virology , Lentivirus/genetics , T-Lymphocyte Subsets/physiology , T-Lymphocyte Subsets/virology , Animals , CD3 Complex/genetics , Cell Line, Tumor , Gene Transfer Techniques , Genetic Engineering/methods , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Hematopoietic Stem Cell Transplantation/methods , Inflammation/genetics , Inflammation/virology , Leukocytes/physiology , Leukocytes/virology , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics
15.
Pol J Vet Sci ; 23(2): 203-209, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32627983

ABSTRACT

The study was carried out in Polish goat population to estimate the prevalence of the nasal cavity infection with various staphylococcal species including methicillin-resistant Staphylococcus aureus(MRSA), investigate the potential permissive role of small ruminant lentivirus (SRLV) infection and determine the level of clonality of S. aureus nasal isolates. Nasal swabs and blood samples were collec-ted from 1300 clinically healthy adult goats from 21 Polish goat herds. Blood samples were serological-ly screened for SRLV. Staphylococci were isolated from nasal swabs and identified using classical microbiological methods, MALDI-TOF, multiplex-PCR, and their clonality was assessed using PFGE. Antimicrobial resistance was determined on the basis of minimum inhibitory concentration and by demonstration of the presence of the mecA gene encoding the multiplex-PCR PBP2a protein and of the five main types of staphylococcal cassette chromosome mec. The apparent prevalence of staphylococ-cal and S. aureus infection of the nasal cavity was 29.1% (CI 95%: 26.9%, 31.5%) and 7.3% (CI 95%: 6.1%, 8.8%), respectively. No relationship was found between the SRLV-infection and the presence of any staphylococcal species including S. aureus (p=0.143). Only 9.8% of S. aureus isolates were resistant to amoxicillin/clavulanic acid and 5.9% to chloramphenicol and ciprofloxacin. All tested isolates proved to be phenotypically and genotypically sensitive to methicillin, which yielded the appar-ent prevalence of MRSA of 0% (CI 95%: 0%, 7.0%). S. aureus isolates show high genetic similarity within goat herds, however vary considerably between herds. Goats do not appear to be an important source of S. aureus for humans in Poland.


Subject(s)
Goat Diseases/microbiology , Lentivirus Infections/veterinary , Nose/microbiology , Staphylococcus/isolation & purification , Animals , Carrier State , Goat Diseases/epidemiology , Goat Diseases/virology , Goats , Lentivirus , Lentivirus Infections/epidemiology , Lentivirus Infections/virology , Staphylococcus/classification
16.
J Vet Diagn Invest ; 32(4): 589-593, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32552528

ABSTRACT

Since 2007, the Autonomous Province of Bolzano-South Tyrol (Italy) has carried out a compulsory eradication program against caprine arthritis encephalitis virus (CAEV) in goats. A drastic seroprevalence reduction was achieved during the initial phase (2007-2011); however, a tailing phenomenon has been observed during the latest years, hampering the achievement of the final goal. CAEV belongs to a group of lentiviruses, called small ruminant lentiviruses (SRLVs), which are antigenically related and can infect both goats and sheep. We investigated the possible link between the tailing phenomenon in goats and the role of sheep as a virus reservoir by comparing serologic results between multispecies farms (where goats and sheep coexist) and monospecies farms (goats only). Goats on multispecies farms had a higher prevalence and seroconversion rate (even if to a rather moderate extent), higher antibody titers, and a higher probability of conclusive results in the genotyping analysis, with more frequent identification of SRLV genotype A (sheep-related) infections. Sheep can serve as a SRLV reservoir, thus contributing to scattered positive tests in goats, causing the tailing phenomenon.


Subject(s)
Arthritis-Encephalitis Virus, Caprine/physiology , Disease Eradication , Disease Reservoirs/veterinary , Goat Diseases/prevention & control , Lentivirus Infections/veterinary , Sheep, Domestic/virology , Animals , Goat Diseases/virology , Goats , Italy , Lentivirus Infections/prevention & control , Lentivirus Infections/virology , Prevalence , Seroconversion
18.
Arch Virol ; 165(7): 1557-1567, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32356187

ABSTRACT

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.


Subject(s)
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
19.
J Dairy Sci ; 103(7): 6407-6411, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32331882

ABSTRACT

Caprine arthritis encephalitis (CAE) is a chronic disease caused by a retrovirus from the Lentivirus genus. No effective vaccines or treatments exist, and therefore genetic selection for CAE resistance might be a feasible alternative. To our best knowledge, no other studies have investigated the genetic architecture of CAE resistance in dairy goats. In this context, this study was designed to estimate genetic parameters for CAE infection in Alpine and Saanen goats using a Bayesian threshold model. A total of 542 adult goats (and >3-generation pedigree), which were group-housed in a population with high CAE prevalence, were tested based on a serological infection assessment test (negative = 1 or positive = 2) and used for this study. Genetic parameters were estimated using the BLUPF90 family programs. There was considerable genetic variability for CAE resistance, and pedigree-based heritability was significantly different from zero (0.026 < heritability < 0.128). Our findings indicate that the prevalence of CAE in goat herds can be reduced or eliminated through direct genetic selection for CAE resistance in addition to proper management strategies.


Subject(s)
Arthritis-Encephalitis Virus, Caprine , Genetic Predisposition to Disease , Goat Diseases/virology , Lentivirus Infections/veterinary , Animals , Bayes Theorem , Goat Diseases/epidemiology , Goats , Lentivirus Infections/genetics , Lentivirus Infections/virology
20.
PLoS One ; 15(3): e0230617, 2020.
Article in English | MEDLINE | ID: mdl-32208446

ABSTRACT

A longitudinal observational study was carried out to evaluate the influence of prenatal exposure to small ruminant lentivirus(SRLV)-infected does on the body weight (BWT) of young kids. The study was carried out in years 2001-2017 in the research dairy goat herd. Goats in the herd were regularly serologically tested and individuals showing clinical signs of caprine arthritis-encephalitis (CAE) were promptly culled. As a result all goats enrolled in the study were asymptomatic. Moreover, kids were weaned immediately after birth, fed on bovine colostrum and kept in strict separation from mothers to prevent SRLV lactogenic transmission. Kids were weighed immediately after birth, and then 1-3 times within the first 3 months of life. In total 620 goat kids were weighed at least once, excluding weighing at birth, providing 992 BWT records. The mixed linear model including four variables fitted as random effects (doe, kid, the year of kid's birth and the exact age of a kid at weighing) and four potential confounders fitted as fixed effects (parity, kid's sex, litter size and birth body weight) was developed and showed that BWT was not significantly associated with SRLV serological status of a doe, regardless of the time for which does had been infected before the delivery of the kid (p = 0.242). This study provides strong evidence that kids born to SRLV-infected does grow equally well as kids from uninfected does, provided that the lactogenic viral transmission is prevented by maintaining strict separation between the offspring and mothers. This observation is important for choosing the most optimal strategy of CAE control in a goat herd.


Subject(s)
Goat Diseases/pathology , Goats/growth & development , Lentivirus Infections/pathology , Animals , Asymptomatic Infections , Body Weight , Female , Goat Diseases/virology , Lentivirus Infections/virology , Linear Models , Longitudinal Studies , Male , Pregnancy , Prenatal Exposure Delayed Effects
SELECTION OF CITATIONS
SEARCH DETAIL
...