Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 452
Filter
1.
Elife ; 102021 07 05.
Article in English | MEDLINE | ID: mdl-34223819

ABSTRACT

Early events in retrovirus transmission are determined by interactions between incoming viruses and frontline cells near entry sites. Despite their importance for retroviral pathogenesis, very little is known about these events. We developed a bioluminescence imaging (BLI)-guided multiscale imaging approach to study these events in vivo. Engineered murine leukemia reporter viruses allowed us to monitor individual stages of retrovirus life cycle including virus particle flow, virus entry into cells, infection and spread for retroorbital, subcutaneous, and oral routes. BLI permitted temporal tracking of orally administered retroviruses along the gastrointestinal tract as they traversed the lumen through Peyer's patches to reach the draining mesenteric sac. Importantly, capture and acquisition of lymph-, blood-, and milk-borne retroviruses spanning three routes was promoted by a common host factor, the I-type lectin CD169, expressed on sentinel macrophages. These results highlight how retroviruses co-opt the immune surveillance function of tissue-resident sentinel macrophages for establishing infection.


Subject(s)
Retroviridae Infections/diagnostic imaging , Retroviridae Infections/transmission , Retroviridae/physiology , Sialic Acid Binding Ig-like Lectin 1/metabolism , Animals , Disease Models, Animal , Female , Humans , Leukemia Virus, Murine , Life Cycle Stages , Lymph Nodes , Macrophages/virology , Male , Mammary Glands, Human/diagnostic imaging , Mammary Glands, Human/virology , Mice , Retroviridae/genetics , Retroviridae Infections/metabolism , Retroviridae Infections/pathology , Sialic Acid Binding Ig-like Lectin 1/genetics , Spleen/diagnostic imaging , Virion , Virus Internalization
2.
OMICS ; 25(6): 358-371, 2021 06.
Article in English | MEDLINE | ID: mdl-34037476

ABSTRACT

About a tenth of all cancers are caused by viruses or associated with viral infection. Recent global events including the coronavirus disease-2019 (COVID-19) pandemic means that human encounter with viruses is increased. Cancer development in individuals with viral infection can take many years after infection, demonstrating that the involvement of viruses in cancer development is a long and complex process. This complexity emanates from individual genetic heterogeneity and the many steps involved in cancer development owing to viruses. The process of tumorigenesis is driven by the complex interaction between several viral factors and host factors leading to the creation of a tumor microenvironment (TME) that is ideal and promotes tumor formation. Viruses associated with human cancers ensure their survival and proliferation through activation of several cellular processes including inflammation, migration, and invasion, resistance to apoptosis and growth suppressors. In addition, most human oncoviruses evade immune detection and can activate signaling cascades including the PI3K-Akt-mTOR, Notch and Wnt pathways associated with enhanced proliferation and angiogenesis. This expert review examines and synthesizes the multiple biological factors related to oncoviruses, and the signaling cascades activated by these viruses contributing to viral oncogenesis. In particular, I examine and review the Epstein-Barr virus, human papillomaviruses, and Kaposi's sarcoma herpes virus in a context of cancer pathogenesis. I conclude with a future outlook on therapeutic targeting of the viruses and their associated oncogenic pathways within the TME. These anticancer strategies can be in the form of, but not limited to, antibodies and inhibitors.


Subject(s)
Epstein-Barr Virus Infections/virology , Neoplasms/virology , Papillomavirus Infections/virology , Retroviridae Infections/virology , Retroviridae/physiology , Sarcoma, Kaposi/virology , Tumor Virus Infections/virology , Alphapapillomavirus/physiology , Carcinogenesis , Cell Transformation, Viral , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/physiology , Herpesvirus 8, Human/physiology , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Neoplasms/therapy , Papillomavirus Infections/pathology , Retroviridae Infections/pathology , Sarcoma, Kaposi/pathology , Signal Transduction , Tumor Microenvironment , Tumor Virus Infections/pathology
3.
Poult Sci ; 100(1): 19-25, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33357681

ABSTRACT

Both reticuloendotheliosis and Marek's disease are neoplastic diseases of chickens caused by reticuloendotheliosis virus (REV) and Marek's disease virus (MDV), respectively. The infection of REV or MDV may lead to clinical tumors and also result in immunosuppression and easily allow secondary infection by other pathogens. Here, we investigated a breeder flock of three-yellow chickens in southern China that had been vaccinated with CVI988/Rispens at hatching and had experienced depression, weakness, reduction in weight gain, and an increased death rate after 120 d of age. The morbidity and mortality were 20% and 10%, respectively, at 140 d of age when this infection was diagnosed. The necropsy of the birds revealed significant tumor-like lesions in the heart, liver, spleen, and ceca. Peripheral blood lymphocytes and tumor-like tissues were sampled for PCR detection and for histopathological observation, for virus isolation and the subsequent immunofluorescent assay on the cell cultures and for gene sequencing of the isolated viruses. A REV isolate GX18NNR1 and a MDV isolate GX18NNM5 were both recovered from the sampled bird. Further phylogenetic analysis based on the env gene of REV and the meq gene of MDV demonstrated that GX18NNR1 was closely related to the reference REV strain MD-2, which was isolated from a contaminated commercial turkey herpesvirus vaccine. In addition, the GX18NNM5 was found to belong to the Chinese very virulent MDV strains' cluster. The coinfection of REV and MDV may contribute to tumor outbreaks with high morbidity and mortality in three-yellow chicken flocks.


Subject(s)
Coinfection , Marek Disease , Neoplasms , Poultry Diseases , Retroviridae Infections , Tumor Virus Infections , Animals , Chickens , China/epidemiology , Coinfection/veterinary , Disease Outbreaks/veterinary , Herpesvirus 2, Gallid/genetics , Marek Disease/epidemiology , Marek Disease/pathology , Neoplasms/etiology , Neoplasms/veterinary , Neoplasms/virology , Phylogeny , Poultry Diseases/epidemiology , Poultry Diseases/pathology , Reticuloendotheliosis virus/classification , Reticuloendotheliosis virus/genetics , Retroviridae Infections/complications , Retroviridae Infections/epidemiology , Retroviridae Infections/pathology , Retroviridae Infections/veterinary , Tumor Virus Infections/complications , Tumor Virus Infections/epidemiology , Tumor Virus Infections/pathology , Tumor Virus Infections/veterinary
4.
BMC Vet Res ; 16(1): 483, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33308224

ABSTRACT

BACKGROUND: Reticuloendotheliosis virus (REV) is a retrovirus that causes severe immunosuppression in poultry. Animals grow slowly under conditions of oxidative stress. In addition, long-term oxidative stress can impair immune function, as well as accelerate aging and death. This study aimed to elucidate the pathogenesis of REV from the perspective of changes in oxidative-antioxidative function following REV infection. METHODS: A total of 80 one-day-old specific pathogen free (SPF) chickens were randomly divided into a control group (Group C) and an REV-infected group (Group I). The chickens in Group I received intraperitoneal injections of REV with 104.62/0.1 mL TCID50. Thymus was collected on day 1, 3, 7, 14, 21, 28, 35, and 49 for histopathology and assessed the status of oxidative stress. RESULTS: In chickens infected with REV, the levels of H2O2 and MDA in the thymus increased, the levels of TAC, SOD, CAT, and GPx1 decreased, and there was a reduction in CAT and Gpx1 mRNA expression compared with the control group. The thymus index was also significantly reduced. Morphological analysis showed that REV infection caused an increase in the thymic reticular endothelial cells, inflammatory cell infiltration, mitochondrial swelling, and nuclear damage. CONCLUSIONS: These results indicate that an increase in oxidative stress enhanced lipid peroxidation, markedly decreased antioxidant function, caused thymus atrophy, and immunosuppression in REV-infected chickens.


Subject(s)
Oxidative Stress , Poultry Diseases/virology , Reticuloendotheliosis virus , Retroviridae Infections/veterinary , Thymus Gland/pathology , Animals , Antioxidants/metabolism , Chickens , Hydrogen Peroxide/metabolism , Poultry Diseases/metabolism , Poultry Diseases/pathology , Retroviridae Infections/metabolism , Retroviridae Infections/pathology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology , Tumor Virus Infections/veterinary
5.
Viruses ; 12(8)2020 08 04.
Article in English | MEDLINE | ID: mdl-32759845

ABSTRACT

Animal retroviruses are known for their transforming potential, and this is also true for the ones hosted by humans, which have gathered expanding attention as one of the potent causative agents in various disease, including specific cancer types. For instance, Human T Lymphotropic virus (HTLV) is a well-studied class of oncoviruses causing T cell leukemia, while human immunodeficiency virus (HIV) leads to acquired immunodeficiency syndrome (AIDS), which is linked to a series of defining cancers including Kaposi sarcoma, certain types of non-Hodgkin lymphoma, and cervical cancer. Of note, in addition to these "modern" exogenous retroviruses, our genome harbors a staggering number of human endogenous retroviruses (HERVs). HERVs are the genetic remnants of ancient retroviral germline infection of human ancestors and are typically silenced in normal tissues due to inactivating mutations and sequence loss. While some HERV elements have been appropriated and contribute to human physiological functions, others can be reactivated through epigenetic dysregulations to express retroviral elements and promote carcinogenesis. Conversely, HERV replication intermediates or protein products can also serve as intrinsic pathogen-associated molecular patterns that cause the immune system to interpret it as an exogenous infection, thereby stimulating immune responses against tumors. As such, HERVs have also been targeted as a potential internal strategy to sensitize tumor cells for promising immunotherapies. In this review, we discuss the dynamic role of human retroviruses in cancer development, focusing on HIV and HERVs contribution. We also describe potential treatment strategies, including immunotherapeutic targeting of HERVs, inhibiting DNA methylation to expose HERV signatures, and the use of antiretroviral drugs against HIV and HERVs, which can be employed as prospective anti-cancer modalities.


Subject(s)
Endogenous Retroviruses/pathogenicity , HIV Infections/virology , Immunotherapy/methods , Neoplasms/virology , Retroviridae Infections/pathology , Animals , Anti-Retroviral Agents/therapeutic use , DNA Methylation , Endogenous Retroviruses/genetics , HIV Infections/complications , HIV Infections/drug therapy , Humans , Mice , Neoplasms/immunology , Prospective Studies
6.
J Biol Chem ; 295(26): 8819-8833, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32385109

ABSTRACT

Retroviral Gag polyproteins are targeted to the inner leaflet of the plasma membrane through their N-terminal matrix (MA) domain. Because retroviruses of different morphogenetic types assemble their immature particles in distinct regions of the host cell, the mechanism of MA-mediated plasma membrane targeting differs among distinct retroviral morphogenetic types. Here, we focused on possible mechanistic differences of the MA-mediated plasma membrane targeting of the B-type mouse mammary tumor virus (MMTV) and C-type HIV-1, which assemble in the cytoplasm and at the plasma membrane, respectively. Molecular dynamics simulations, together with surface mapping, indicated that, similarly to HIV-1, MMTV uses a myristic switch to anchor the MA to the membrane and electrostatically interacts with phosphatidylinositol 4,5-bisphosphate to stabilize MA orientation. We observed that the affinity of MMTV MA to the membrane is lower than that of HIV-1 MA, possibly related to their different topologies and the number of basic residues in the highly basic MA region. The latter probably reflects the requirement of C-type retroviruses for tighter membrane binding, essential for assembly, unlike for D/B-type retroviruses, which assemble in the cytoplasm. A comparison of the membrane topology of the HIV-1 MA, using the surface-mapping method and molecular dynamics simulations, revealed that the residues at the HIV-1 MA C terminus help stabilize protein-protein interactions within the HIV-1 MA lattice at the plasma membrane. In summary, HIV-1 and MMTV share common features such as membrane binding of the MA via hydrophobic interactions and exhibit several differences, including lower membrane affinity of MMTV MA.


Subject(s)
Cell Membrane/metabolism , HIV Infections/metabolism , HIV-1/physiology , Mammary Tumor Virus, Mouse/physiology , Retroviridae Infections/metabolism , Tumor Virus Infections/metabolism , Animals , Cell Membrane/pathology , HIV Infections/pathology , Host-Pathogen Interactions , Humans , Mice , Models, Molecular , Retroviridae Infections/pathology , Tumor Virus Infections/pathology , Virus Assembly
7.
J Vet Diagn Invest ; 32(3): 454-457, 2020 May.
Article in English | MEDLINE | ID: mdl-32274976

ABSTRACT

An 8-y-old castrated male, outdoor European shorthair cat was presented with a history of hindlimb weakness and paralysis. Disease progression was continuous from the onset; deep algesia disappeared at the final stage. Radiography of the vertebral column was unremarkable; along with patient history and physical examination results, magnetic resonance imaging suggested inflammatory lesions in the spinal cord, although neoplasia could not be ruled out. Feline leukemia virus (FeLV) positivity was confirmed by a serum ELISA prior to euthanasia. Upon postmortem examination, hemorrhages were present in the spinal cord at the level of vertebrae T7-8. Histologic and immunohistochemical analysis revealed primary diffuse large B-cell lymphoma of the spinal cord with multifocal myelomalacia and hemorrhages. To determine the presence of a pathogen within the lesion, we developed a novel in situ hybridization protocol for FeLV (RNAscope). The reaction revealed large amounts of FeLV viral RNA in the tumor cells.


Subject(s)
Cat Diseases/virology , In Situ Hybridization/veterinary , Leukemia Virus, Feline/genetics , Lymphoma, B-Cell/veterinary , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Cat Diseases/pathology , Cats , Leukemia Virus, Feline/physiology , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/virology , Male , RNA, Viral/analysis , RNA, Viral/isolation & purification , Retroviridae Infections/pathology , Retroviridae Infections/virology , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
8.
PLoS Pathog ; 16(3): e1008340, 2020 03.
Article in English | MEDLINE | ID: mdl-32226027

ABSTRACT

Combination immunotherapy (CIT) is currently applied as a treatment for different cancers and is proposed as a cure strategy for chronic viral infections. Whether such therapies are efficient during an acute infection remains elusive. To address this, inhibitory receptors were blocked and regulatory T cells depleted in acutely Friend retrovirus-infected mice. CIT resulted in a dramatic expansion of cytotoxic CD4+ and CD8+ T cells and a subsequent reduction in viral loads. Despite limited viral replication, mice developed fatal immunopathology after CIT. The pathology was most severe in the gastrointestinal tract and was mediated by granzyme B producing CD4+ and CD8+ T cells. A similar post-CIT pathology during acute Influenza virus infection of mice was observed, which could be prevented by vaccination. Melanoma patients who developed immune-related adverse events under immune checkpoint CIT also presented with expanded granzyme-expressing CD4+ and CD8+ T cell populations. Our data suggest that acute infections may induce immunopathology in patients treated with CIT, and that effective measures for infection prevention should be applied.


Subject(s)
Antibodies/administration & dosage , Melanoma/immunology , Melanoma/therapy , Retroviridae Infections/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Virus Infections/immunology , Animals , B7-H1 Antigen/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Friend murine leukemia virus/physiology , Humans , Immunotherapy/adverse effects , Melanoma/pathology , Mice , Mice, Inbred C57BL , Retroviridae Infections/pathology , Retroviridae Infections/virology , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
9.
J Vet Diagn Invest ; 32(3): 389-393, 2020 May.
Article in English | MEDLINE | ID: mdl-32233842

ABSTRACT

Reticuloendotheliosis virus (REV) can cause runting, immunosuppression, acute reticulum cell neoplasia, and chronic lymphoid tumors in a variety of domestic and wild birds. We diagnosed a case of reticuloendotheliosis with obvious tumors in liver and kidney. We isolated and sequenced the virus and performed pathogenicity testing of the REV strain. Immunohistochemistry and PCR confirmed that the diseased layer chickens were infected with REV. The strain, named BJ1503, was successfully isolated from the case by inoculation of tissue homogenates onto chicken embryo fibroblasts. The length of the proviral REV genome is 8,293 nucleotides. The isolate had 99.7% identity with REV-HA9901 (AY842951.1), which was isolated from Jiangsu, China, in 1999. The chickens infected with REV-BJ1503 had depressed weight gain and lymphoid atrophy. Our findings suggest that REV isolate BJ1503 was phylogenetically close to the earlier strain found in China, with minor variations, and the virus was associated with severe production problems.


Subject(s)
Chickens , Poultry Diseases/virology , Reticuloendotheliosis Viruses, Avian/isolation & purification , Reticuloendotheliosis Viruses, Avian/pathogenicity , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , China , Female , Phylogeny , Poultry Diseases/pathology , Retroviridae Infections/pathology , Retroviridae Infections/virology , Sequence Analysis, RNA/veterinary , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Virulence
10.
Sci Rep ; 10(1): 263, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31937823

ABSTRACT

Koala retrovirus (KoRV) displays features of both an endogenous and exogenous virus and is linked to neoplasia and immunosuppression in koalas. This study explores the apparent differences in the nature and impact of KoRV infection between geographically and genetically separated "northern" and "southern" koala populations, by investigating the disease status, completeness of the KoRV genome and the proviral (DNA) and viral (RNA) loads of 71 northern and 97 southern koalas. All northern animals were positive for all KoRV genes (gag, pro-pol and env) in both DNA and RNA forms, whereas many southern animals were missing one or more KoRV genes. There was a significant relationship between the completeness of the KoRV genome and clinical status in this population. The proviral and viral loads of the northern population were significantly higher than those of the southern population (P < 0.0001), and many provirus-positive southern animals failed to express any detectable KoRV RNA. Across both populations there was a positive association between proviral load and neoplasia (P = 0.009). Potential reasons for the differences in the nature of KoRV infection between the two populations are discussed.


Subject(s)
Retroviridae Infections/pathology , Retroviridae/genetics , Aging/genetics , Animals , Australia/epidemiology , DNA/metabolism , Female , Gene Products, env/genetics , Gene Products, env/metabolism , Gene Products, gag/genetics , Gene Products, gag/metabolism , Gene Products, pol/genetics , Gene Products, pol/metabolism , Male , Phascolarctidae , Proviruses/genetics , RNA, Viral/blood , Retroviridae/isolation & purification , Retroviridae Infections/epidemiology , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Viral Load
11.
J Vet Diagn Invest ; 31(5): 761-765, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31378167

ABSTRACT

We investigated the histologic findings and viral antigen distribution in 3 cases of natural coinfection of layer hens with subgroup J avian leukosis virus (ALV-J), Marek's disease virus (MDV), and reticuloendotheliosis virus (REV) in hens. At autopsy, diseased hens were found to have hepatosplenomegaly and thickened proventriculi, with white tumor nodules in the liver, spleen, lung, kidney, and ovary. Microscopically, most tissues had been infiltrated by neoplastic lymphocytes; the spleen, lung, proventriculus, heart, and liver had been infiltrated by both neoplastic lymphocytes and myeloblastic cells and/or primitive reticular cells. Fluorescence multiplex immunohistochemistry staining revealed ALV-J, MDV, and REV antigens co-expressed in the same tissue, even the same cell.


Subject(s)
Avian Leukosis/virology , Chickens , Coinfection/veterinary , Marek Disease/virology , Poultry Diseases/virology , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Antigens, Viral/analysis , Avian Leukosis/immunology , Avian Leukosis/pathology , Avian Leukosis Virus/physiology , Coinfection/immunology , Coinfection/pathology , Coinfection/virology , Female , Herpesvirus 2, Gallid/physiology , Marek Disease/immunology , Marek Disease/pathology , Poultry Diseases/immunology , Poultry Diseases/pathology , Reticuloendotheliosis virus/physiology , Retroviridae Infections/immunology , Retroviridae Infections/pathology , Retroviridae Infections/virology , Tumor Virus Infections/immunology , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
12.
J Gen Virol ; 100(2): 266-277, 2019 02.
Article in English | MEDLINE | ID: mdl-30608228

ABSTRACT

Recently, a large number of Japanese macaques (Macaca fuscata) died of an unknown hemorrhagic syndrome at Kyoto University Primate Research Institute (KUPRI) and an external breeding facility for National Institute for Physiological Sciences (NIPS). We previously reported that the hemorrhagic syndrome of Japanese macaques at KUPRI was caused by infection with simian retrovirus 4 (SRV-4); however, the cause of similar diseases that occurred at the external breeding facility for NIPS was still unknown. In this study, we isolated SRV-5 from Japanese macaques exhibiting thrombocytopenia and then constructed an infectious molecular clone of the SRV-5 isolate. When the SRV-5 isolate was inoculated into two Japanese macaques, severe thrombocytopenia was induced in one of two macaques within 22 days after inoculation. Similarly, the clone-derived virus was inoculated into the other two Japanese macaques, and one of two macaques developed severe thrombocytopenia within 22 days. On the other hand, the remaining two of four macaques survived as asymptomatic carriers even after administering an immunosuppressive agent, dexamethasone. As determined by real-time PCR, SRV-5 infected a variety of tissues in Japanese macaques, especially in digestive and lymph organs. We also identified the SRV-5 receptor as ASCT2, a neutral amino acid transporter in Japanese macaques. Taken together, we conclude that the causative agent of hemorrhagic syndrome occurred at the external breeding facility for NIPS was SRV-5.


Subject(s)
Hemorrhagic Disorders/veterinary , Monkey Diseases/pathology , Monkey Diseases/virology , Retroviridae Infections/veterinary , Retroviruses, Simian/growth & development , Retroviruses, Simian/pathogenicity , Thrombocytopenia/veterinary , Animals , Hemorrhagic Disorders/pathology , Hemorrhagic Disorders/virology , Macaca , Retroviridae Infections/pathology , Retroviridae Infections/virology , Retroviruses, Simian/isolation & purification , Thrombocytopenia/pathology , Thrombocytopenia/virology
13.
Open Vet J ; 9(3): 246-252, 2019 10.
Article in English | MEDLINE | ID: mdl-31998618

ABSTRACT

Background: Cerebellar cortical degeneration (CCD) is the premature death of cerebellar neurons of heterogeneous etiology that is uncommonly observed as a neurological complication of certain neoplasia. Case Description: Here, we report an 8-month-old male domestic cat with altered consciousness, symmetric ataxia, hypermetric gait, vertical positional nystagmus, mydriasis, strabismus, intention tremor of the head, and increased patellar reflexes. Neuroanatomical diagnosis suggested a multifocal brain dysfunction (cerebellar and cerebral). The cat tested seropositive for feline leukemia virus. Cerebrospinal fluid analysis indicated mononuclear and neutrophilic pleocytosis. Contrast computed tomography imaging revealed multiple hypodense heterogeneous areas in both cerebral hemispheres, mild ventriculomegaly at the level of the caudal fossa, and a circular sharply marginated, homogeneously hyperdense mass occupying the right cerebellar hemisphere. Postmortem study indicated a 1.1 × 1.3 × 1.2 cm mass in the right cerebellar hemisphere close to the vermis. Histopathological analysis showed diffuse and severe Purkinje cell loss with a decrease in granular cell density and moderate gliosis compatible with CCD. Further, numerous neoplastic lymphoid cells were observed in the infiltrated mass, consistent with a diagnosis of central nervous system (CNS) lymphoma. Immunohistochemistry showed CD20 expression, indicative of a B-cell immunophenotype. In humans, CCD is reported as a rare paraneoplastic syndrome in patients with Hodgkin lymphoma. CNS lymphoma and/or Feline Leukemia Virus (FeLV) infection were both considered as a possible cause of CCD in this case. Conclusion: This is the first described case of possible paraneoplastic cerebellar cortical degeneration associated with CNS lymphoma and/or FeLV infection in a domestic cat.


Subject(s)
Cat Diseases/virology , Leukemia Virus, Feline/physiology , Paraneoplastic Cerebellar Degeneration/veterinary , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Animals , Cat Diseases/pathology , Cats , Cerebellum/pathology , Male , Paraneoplastic Cerebellar Degeneration/pathology , Paraneoplastic Cerebellar Degeneration/virology , Retroviridae Infections/pathology , Retroviridae Infections/virology , Tumor Virus Infections/pathology , Tumor Virus Infections/virology
14.
Front Immunol ; 9: 1947, 2018.
Article in English | MEDLINE | ID: mdl-30210499

ABSTRACT

Natural killer (NK) cells play a key role in host defense against cancer and viral infections. It was shown that NK cells are important for the control of acute retroviral infections, but their antiviral activity depends on multiple parameters such as viral inoculation dose, interactions with myeloid cell types and the cytokine milieu. In addition, during an ongoing retroviral infection regulatory T cells (Tregs) can suppress NK cell functions. However, the precise role of Tregs on the initial NK cell response and their immediate antiviral activity after an acute retroviral infection is still unknown. Here we show that thymus-derived Tregs suppress the proliferation, effector functions and cytotoxicity of NK cells very early during acute Friend Retrovirus (FV) infection. Tregs exhibited an activated phenotype and increased the production of the immunosuppressive cytokines IL-10 and TGF-ß after FV infection of mice. Neutralization of the immunosuppressive cytokine IL-10 resulted in a significant augmentation of NK cell functions. Although the activation of dendritic cells (DCs) and macrophages as well as the IL-15 cytokine levels were increased after Treg depletion, Tregs mainly affect the NK cell activity in an IL-10-regulated pathway. In this study we demonstrate an IL-10-dependent suppression of NK cells by activated Tregs during the first days of a retroviral infection.


Subject(s)
Friend murine leukemia virus/immunology , Immunity, Cellular , Interleukin-10/immunology , Killer Cells, Natural/immunology , Retroviridae Infections/immunology , T-Lymphocytes, Regulatory/immunology , Acute Disease , Animals , Dendritic Cells/immunology , Dendritic Cells/pathology , Female , Killer Cells, Natural/pathology , Male , Mice , Retroviridae Infections/pathology , T-Lymphocytes, Regulatory/pathology
15.
J Neuroimmune Pharmacol ; 13(3): 279-291, 2018 09.
Article in English | MEDLINE | ID: mdl-29656370

ABSTRACT

Retroviruses comprise an ancient and varied group of viruses with the unique ability to integrate DNA from an RNA transcript into the genome, a subset of which are able to integrate in humans. The timing of these integrations during human history has dictated whether these viruses have remained exogenous and given rise to various human diseases or have become inseparable from the host genome (endogenous retroviruses). Given the ability of retroviruses to integrate into the host and subsequently co-opt host cellular process for viral propagation, retroviruses have been shown to be closely associated with several cellular processes including exosome formation. Exosomes are 30-150 nm unilamellar extracellular vesicles that originate from intraluminal vesicles (ILVs) that form in the endosomal compartment. Exosomes have been shown to be important in intercellular communication and immune cell function. Almost every cell type studied has been shown to produce these types of vesicles, with the cell type dictating the contents, which include proteins, mRNA, and miRNAs. Importantly, recent evidence has shown that infection by viruses, including retroviruses, alter the contents and subsequent function of produced exosomes. In this review, we will discuss the important retroviruses associated with human health and disease. Furthermore, we will delve into the impact of exosome formation and manipulation by integrated retroviruses on human health, survival, and human retroviral disease pathogenesis.


Subject(s)
Exosomes/pathology , Retroviridae Infections/pathology , Animals , HIV Infections/pathology , HIV Infections/virology , Humans , Retroviridae , Retroviridae Infections/virology
16.
J Infect Dis ; 218(1): 144-151, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29608711

ABSTRACT

Background: A spillover of simian foamy virus (SFV) to humans, following bites from infected nonhuman primates (NHPs), is ongoing in exposed populations. These retroviruses establish persistent infections of unknown physiological consequences to the human host. Methods: We performed a case-control study to compare 24 Cameroonian hunters infected with gorilla SFV and 24 controls matched for age and ethnicity. A complete physical examination and blood test were performed for all participants. Logistic regression and Wilcoxon signed rank tests were used to compare cases and controls. Results: The cases had significantly lower levels of hemoglobin than the controls (median, 12.7 vs 14.4 g/dL; P = .01). Basophil levels were also significantly lower in cases than controls, with no differences for other leukocyte subsets. Cases had significantly higher urea, creatinine, protein, creatinine phosphokinase, and lactate dehydrogenase levels and lower bilirubin levels than controls. Cases and controls had similar frequencies of general, cutaneous, gastrointestinal, neurological, and cardiorespiratory signs. Conclusions: The first case-control study of apparently healthy SFV-infected Cameroonian hunters showed the presence of hematological abnormalities. A thorough clinical and laboratory workup is now needed to establish the medical relevance of these observations because more than half of cases had mild or moderate anemia. Clinical Trials Registration: NCT03225794.


Subject(s)
Retroviridae Infections/pathology , Simian foamy virus/isolation & purification , Adult , Aged , Animals , Basophils/immunology , Blood Chemical Analysis , Cameroon , Case-Control Studies , Humans , Male , Middle Aged , Retroviridae Infections/virology , Young Adult
17.
Arch Virol ; 163(6): 1663-1669, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29460003

ABSTRACT

Over a period of 7 years (2004-2011), samples from 34 diseased reptiles provided by local governments, zoos, and pet shops were tested for viral infection. Animals were diagnosed based on clinical signs, including loss of appetite, diarrhea, rhinorrhea, and unexpected sudden death. Most of the exotic animals had gastrointestinal problems, such as mucosal redness and ulcers, while the native animals had no clinical symptoms. Viral sequences were found in seven animals. Retroviral genes were amplified from samples from five Burmese pythons (Python molurus bivittatus), an adenovirus was detected in a panther chameleon (Furcifer pardalis), and an adenovirus and a paramyxovirus were detected in a tropical girdled lizard (Cordylus tropidosternum). Phylogenetic analysis of retroviruses and paramyxoviruses showed the highest sequence identity to both a Python molurus endogenous retrovirus and a Python curtus endogenous retrovirus and to a lizard isolate, respectively. Partial sequencing of an adenoviral DNA polymerase gene from the lizard isolate suggested that the corresponding virus was a novel isolate different from the reference strain (accession no. AY576677.1). The virus was not isolated but was detected, using molecular genetic techniques, in a lizard raised in a pet shop. This animal was also coinfected with a paramyxovirus.


Subject(s)
Adenoviridae/genetics , DNA-Directed DNA Polymerase/genetics , Paramyxoviridae/genetics , Phylogeny , Reptiles/virology , Retroviridae/genetics , Viral Proteins/genetics , Adenoviridae/classification , Adenoviridae/isolation & purification , Adenoviridae/pathogenicity , Adenoviridae Infections/mortality , Adenoviridae Infections/pathology , Adenoviridae Infections/veterinary , Adenoviridae Infections/virology , Animals , DNA, Viral/genetics , Paramyxoviridae/classification , Paramyxoviridae/isolation & purification , Paramyxoviridae/pathogenicity , Paramyxoviridae Infections/mortality , Paramyxoviridae Infections/pathology , Paramyxoviridae Infections/veterinary , Paramyxoviridae Infections/virology , Republic of Korea , Retroviridae/classification , Retroviridae/isolation & purification , Retroviridae/pathogenicity , Retroviridae Infections/mortality , Retroviridae Infections/pathology , Retroviridae Infections/veterinary , Retroviridae Infections/virology
18.
PLoS Pathog ; 14(2): e1006776, 2018 02.
Article in English | MEDLINE | ID: mdl-29447279

ABSTRACT

Tight regulation of immune responses is not only critical for preventing autoimmune diseases but also for preventing immunopathological damage during infections in which overactive immune responses may be more harmful for the host than the pathogen itself. Regulatory T cells (Tregs) play a critical role in this regulation, which was discovered using the Friend retrovirus (FV) mouse model. Subsequent FV studies revealed basic biological information about Tregs, including their suppressive activity on effector cells as well as the molecular mechanisms of virus-induced Treg expansion. Treg suppression not only limits immunopathology but also prevents complete elimination of pathogens contributing to chronic infections. Therefore, Tregs play a complex role in the pathogenesis of persistent retroviral infections. New therapeutic concepts to reactivate effector T-cell responses in chronic viral infections by manipulating Tregs also came from work with the FV model. This knowledge initiated many studies to characterize the role of Tregs in HIV pathogenesis in humans, where a complex picture is emerging. On one hand, Tregs suppress HIV-specific effector T-cell responses and are themselves targets of infection, but on the other hand, Tregs suppress HIV-induced immune hyperactivation and thus slow the infection of conventional CD4+ T cells and limit immunopathology. In this review, the basic findings from the FV mouse model are put into perspective with clinical and basic research from HIV studies. In addition, the few Treg studies performed in the simian immunodeficiency virus (SIV) monkey model will also be discussed. The review provides a comprehensive picture of the diverse role of Tregs in different retroviral infections and possible therapeutic approaches to treat retroviral chronicity and pathogenesis by manipulating Treg responses.


Subject(s)
Host-Pathogen Interactions , Models, Immunological , Retroviridae Infections/immunology , Retroviridae/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Humans , Immune Tolerance , Immunosuppression Therapy , Lymphopoiesis , Retroviridae/physiology , Retroviridae Infections/pathology , Retroviridae Infections/therapy , Retroviridae Infections/virology , T-Lymphocytes, Regulatory/pathology , T-Lymphocytes, Regulatory/virology
19.
Int J Cancer ; 142(5): 976-987, 2018 03 01.
Article in English | MEDLINE | ID: mdl-28975621

ABSTRACT

The antitumor effectiveness of cyclophosphamide (CTX) and other chemotherapeutics was shown to rely not only on direct cytotoxicity but also on immunogenic tumor cell death and systemic immunomodulatory mechanisms, including regulatory T cell (Treg) depletion, Th1 cell polarization, type I interferon (IFN) and proinflammatory cytokine production. IFN regulatory factor (IRF)-1 is a transcriptional regulator of IFNs and IFN-inducible genes, involved in the control of Th1 and Treg differentiation and in sterile inflammation. Aim of this study was to explore the role of IRF-1 in CTX-induced antitumor effects and related immune activities. This study shows for the first time that IRF-1 is important for the antitumor efficacy of CTX in mice. Moreover, experiments in tumor-bearing C57BL/6 mice showed that Irf1 gene expression in the spleen was transiently increased following CTX administration and correlated with the induction of Th1 cell expansion and of Il12p40 gene expression, which is the main Th1-driving cytokine. At the same time, CTX administration reduced both Foxp3 expression and Treg cell percentages. These effects were abrogated in Irf1-/- mice. Further experiments showed that the gene and/or protein expression of caspase-1, iNOS, IL-1ß, IL-6 and CXCL10 and the levels of nitric oxide were modulated following CTX in an IRF-1-direct- or -indirect-dependent manner, and highlighted the importance of caspase-1 in driving the sterile inflammatory response to CTX. Our data identify IRF-1 as important for the antitumor efficacy of CTX and for the regulation of many immunomodulatory activities of CTX, such as Th1 polarization, Treg depletion and inflammation.


Subject(s)
Cyclophosphamide/pharmacology , Inflammasomes/immunology , Interferon Regulatory Factor-1/physiology , Leukemia, Experimental/drug therapy , Retroviridae Infections/drug therapy , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Tumor Virus Infections/drug therapy , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Cytokines/metabolism , Female , Gene Expression Regulation/drug effects , Leukemia, Experimental/immunology , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Rauscher Virus/pathogenicity , Retroviridae Infections/immunology , Retroviridae Infections/metabolism , Retroviridae Infections/pathology , Tumor Cells, Cultured , Tumor Virus Infections/immunology , Tumor Virus Infections/metabolism , Tumor Virus Infections/pathology
20.
Retrovirology ; 14(1): 55, 2017 12 02.
Article in English | MEDLINE | ID: mdl-29197389

ABSTRACT

BACKGROUND: Foamy viruses (FV) are ancient complex retroviruses that differ from orthoretroviruses such as human immunodeficiency virus (HIV) and murine leukemia virus (MLV) and comprise a distinct subfamily of retroviruses, the Spumaretrovirinae. FV are ubiquitous in their natural hosts, which include cows, cats, and nonhuman primates (NHP). FV are transmitted mainly through saliva and appear nonpathogenic by themselves, but they may increase morbidity of other pathogens in coinfections. CONCLUSIONS: This review summarizes and discusses what is known about FV infection of natural hosts. It also emphasizes what is known about FV zoonotic infections A large number of studies have revealed that the FV of NHP, simian foamy viruses (SFV), are transmitted to humans who interact with infected NHP. SFV from a variety of NHP establish persistent infection in humans, while bovine foamy virus and feline foamy virus rarely or never do. The possibility of FV recombination and mutation leading to pathogenesis is considered. Since humans can be infected by SFV, a seemingly nonpathogenic virus, there is interest in using SFV vectors for human gene therapy. In this regard, detailed understanding of zoonotic SFV infection is highly relevant.


Subject(s)
Retroviridae Infections/transmission , Spumavirus , Zoonoses/virology , Animals , Coinfection , Humans , Retroviridae Infections/pathology , Retroviridae Infections/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...