Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Methods Mol Biol ; 2808: 153-165, 2024.
Article in English | MEDLINE | ID: mdl-38743369

ABSTRACT

Domestic cats are the natural host of feline morbilliviruses (FeMV). Although other species can also be infected (such as dogs and opossums), no laboratory animal infection model is established so far. In vitro models for studying the molecular pathogenesis are therefore needed. For this purpose, propagation and titration of FeMV are key techniques. Unlike other morbilliviruses, such as canine distemper virus (CDV) or measles virus (MV), FeMV is a slow growing virus in cell culture and is difficult to titrate using classical plaque techniques. Here we describe methods for the efficient isolation of FeMV from natural sources (e.g., urine), the propagation of viral stocks, and their titration. In addition, we establish the generation of a three-dimensional infection model mimicking the feline tubular epithelium.


Subject(s)
Morbillivirus Infections , Morbillivirus , Animals , Cats , Morbillivirus/pathogenicity , Morbillivirus/genetics , Morbillivirus/physiology , Morbillivirus Infections/veterinary , Morbillivirus Infections/virology , Kidney/virology , Kidney/cytology , Cat Diseases/virology , Cells, Cultured , Virus Cultivation/methods , Disease Models, Animal , Primary Cell Culture/methods
2.
Vet Microbiol ; 260: 109163, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34311269

ABSTRACT

Small ruminant morbillivirus (SRMV) is a highly contagious and economically important viral disease of small domestic and wild ruminants. Difficulty with its stable proliferation in ovis aries-derived cells has led to a relative lag in the study of its natural immunity and pathogenesis. Here we report the antiviral properties of ZAP against SRMV, a single-stranded negative-stranded RNA virus of the genus Morbillivirus. ZAP expression was significantly induced in sheep endometrial epithelial cells following SRMV infection. ZAP inhibited SRMV replication in cells after infection, while its overexpression in Vero-SLAM cells significantly increased their resistance to SRMV replication. The ZAP protein co-localized with SRMV RNA in the cytoplasm and ZAP-responsive elements were mapped to the 5' untranslated region of SRMV nucleocapsid, phosphoprotein, matrix, and fusion. In summary, ZAP confers resistance to SRMV infection by directly targeting viral RNA and inhibiting viral replication. Our findings further extend the ranges of viral targets of ZAP and help elucidate the mechanism of SRMV replication.


Subject(s)
Morbillivirus Infections/veterinary , Morbillivirus/physiology , RNA-Binding Proteins/metabolism , Animals , Chlorocebus aethiops , Endometrium/virology , Epithelial Cells/virology , Female , HEK293 Cells , Humans , Morbillivirus Infections/virology , RNA, Viral/genetics , RNA-Binding Proteins/genetics , Sheep , Vero Cells , Virus Replication
3.
Viruses ; 13(4)2021 04 15.
Article in English | MEDLINE | ID: mdl-33921104

ABSTRACT

Feline morbillivirus (FeMV) was identified for the first time in stray cats in 2012 in Hong Kong and, since its discovery, it was reported in domestic cats worldwide. Although a potential association between FeMV infection and tubulointerstitial nephritis (TIN) has been suggested, this has not been proven, and the subject remains controversial. TIN is the most frequent histopathological finding in the context of feline chronic kidney disease (CKD), which is one of the major clinical pathologies in feline medicine. FeMV research has mainly focused on defining the epidemiology, the role of FeMV in the development of CKD, and its in vitro tropism, but the pathogenicity of FeMV is still not clear, partly due to its distinctive biological characteristics, as well as to a lack of a cell culture system for its rapid isolation. In this review, we summarize the current knowledge of FeMV infection, including genetic diversity of FeMV strains, epidemiology, pathogenicity, and clinicopathological findings observed in naturally infected cats.


Subject(s)
Cat Diseases , Morbillivirus Infections , Morbillivirus/physiology , Renal Insufficiency, Chronic , Animals , Cat Diseases/pathology , Cat Diseases/virology , Cats , Kidney/virology , Morbillivirus Infections/epidemiology , Morbillivirus Infections/veterinary , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/veterinary
4.
Viruses ; 13(3)2021 02 25.
Article in English | MEDLINE | ID: mdl-33669141

ABSTRACT

Viruses are highly dependent on the host they infect. Their dependence triggers processes of virus-host co-adaptation, enabling viruses to explore host resources whilst escaping immunity. Scientists have tackled viral-host interplay at differing levels of complexity-in individual hosts, organs, tissues and cells-and seminal studies advanced our understanding about viral lifecycles, intra- or inter-species transmission, and means to control infections. Recently, it emerged as important to address the physical properties of the materials in biological systems; membrane-bound organelles are only one of many ways to separate molecules from the cellular milieu. By achieving a type of compartmentalization lacking membranes known as biomolecular condensates, biological systems developed alternative mechanisms of controlling reactions. The identification that many biological condensates display liquid properties led to the proposal that liquid-liquid phase separation (LLPS) drives their formation. The concept of LLPS is a paradigm shift in cellular structure and organization. There is an unprecedented momentum to revisit long-standing questions in virology and to explore novel antiviral strategies. In the first part of this review, we focus on the state-of-the-art about biomolecular condensates. In the second part, we capture what is known about RNA virus-phase biology and discuss future perspectives of this emerging field in virology.


Subject(s)
Host-Pathogen Interactions/physiology , Virus Physiological Phenomena , Animals , Biophysical Phenomena , HIV/physiology , Humans , Influenza A virus/physiology , Morbillivirus/physiology , Organelles/virology , SARS-CoV-2/physiology , Vesiculovirus/physiology , Virus Diseases/virology , Virus Internalization
5.
Diabetes Metab Syndr ; 14(6): 1783-1789, 2020.
Article in English | MEDLINE | ID: mdl-32947109

ABSTRACT

BACKGROUND AND AIMS: Type 1 diabetes (T1D) is a multifactorial autoimmune disease that combines genetics and environmental factors. The aim of this study is to determine the environmental risk factors and to investigate how virals infections are risks factors for type 1 diabetics whom have HLA DR3/DR4 predisposition in our population. METHODS: This study includes 233 subjects, 145 diabetics and 88 controls from regions of the extreme western of Algeria. All the informations related to the disease were collected using predesigned questionnaire. Using in silico approach, we attempt to improve the understanding of this analytical result by molecular mimicry, which is associated with the breakdown of several autoimmune pathologies. RESULTS: The statistical study showed that history of varicella and measles infection and T1D related inheritance and type 2 diabetes are risk factors for T1D in the population of Tlemcen. We have determined the homologous antigenic regions between the glycoprotein "gE" of the varicella virus, the "hemagglutinin" of measles and the human protein "HSP60" at the level of their sequence and 3D structure. These cross-reactive epitopes bind to MHC class II molecules (HLA DR3/DR4) that predispose to T1D but not to MHC class II molecules (HLA DR2) that protect against T1D. This epitopes induce Th2 cells but only "hemagglutinin" and "Hsp60" can activate Th1 differentiation. This indicates their potential to destroy pancreatic cells ß. CONCLUSION: Our study can allow us to adapt biological markers to genetically predisposed T1D and to establish a preventive strategy for healthy genetic predisposed individuals in Tlemcen population.


Subject(s)
Diabetes Mellitus, Type 1/epidemiology , HLA-DR3 Antigen/metabolism , HLA-DR4 Antigen/metabolism , Measles/complications , Molecular Mimicry , Varicella Zoster Virus Infection/complications , Algeria/epidemiology , Biomarkers/analysis , Blood Glucose/analysis , Case-Control Studies , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/virology , Follow-Up Studies , HLA-DR3 Antigen/immunology , Herpesvirus 3, Human/physiology , Humans , Morbillivirus/physiology , Prognosis
6.
Curr Opin Virol ; 41: 38-45, 2020 04.
Article in English | MEDLINE | ID: mdl-32344228

ABSTRACT

Like measles virus (MV), whose primary hosts are humans, non-human animal morbilliviruses use SLAM (signaling lymphocytic activation molecule) and PVRL4 (nectin-4) expressed on immune and epithelial cells, respectively, as receptors. PVRL4's amino acid sequence is highly conserved across species, while that of SLAM varies significantly. However, non-host animal SLAMs often function as receptors for different morbilliviruses. Uniquely, human SLAM is somewhat specific for MV, but canine distemper virus, which shows the widest host range among morbilliviruses, readily gains the ability to use human SLAM. The host range for morbilliviruses is also modulated by their ability to counteract the host's innate immunity, but the risk of cross-species transmission of non-human animal morbilliviruses to humans could occur if MV is successfully eradicated.


Subject(s)
Morbillivirus Infections/veterinary , Morbillivirus Infections/virology , Morbillivirus/physiology , Viral Zoonoses/transmission , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Host Specificity , Humans , Morbillivirus/genetics , Morbillivirus Infections/metabolism , Morbillivirus Infections/transmission , Receptors, Virus/genetics , Receptors, Virus/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1/genetics , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Viral Zoonoses/genetics , Viral Zoonoses/metabolism , Viral Zoonoses/virology
7.
Microbiol Immunol ; 64(8): 578-583, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32215955

ABSTRACT

Morbilliviruses use the signaling lymphocyte activation molecule (SLAM) as a receptor to infect their hosts. Seals are almost the only animal species that show apparent infection with phocine distemper virus (PDV). Seal SLAM functioned as a PDV receptor. However, dolphin- and dog-SLAM molecules, but not human SLAM, were also fully functional PDV receptors. These data suggest that the host range of PDV is not simply determined by its SLAM usage. However, human nonsusceptibility to PDV infection may be at least partly attributable to the inability of PDV to use human SLAM as a receptor.


Subject(s)
Distemper Virus, Canine/physiology , Distemper Virus, Phocine/physiology , Morbillivirus/physiology , Receptors, Virus/physiology , Signaling Lymphocytic Activation Molecule Family Member 1/physiology , Animals , Cell Line , Chlorocebus aethiops , Distemper/virology , Dogs/virology , Humans , Phoca/virology , Receptors, Virus/genetics , Signaling Lymphocytic Activation Molecule Family Member 1/genetics , Stenella/virology , Vero Cells
8.
Viruses ; 11(10)2019 10 14.
Article in English | MEDLINE | ID: mdl-31615092

ABSTRACT

Canine distemper virus (CDV) and phocine distemper (PDV) are closely-related members of the Paramyxoviridae family, genus morbillivirus, in the order Mononegavirales. CDV has a broad host range among carnivores. PDV is thought to be derived from CDV through contact between terrestrial carnivores and seals. PDV has caused extensive mortality in Atlantic seals and other marine mammals, and more recently has spread to the North Pacific Ocean. CDV also infects marine carnivores, and there is evidence of morbillivirus infection of seals and other species in Antarctica. Recently, CDV has spread to felines and other wildlife species in the Serengeti and South Africa. Some CDV vaccines may also have caused wildlife disease. Changes in the virus haemagglutinin (H) protein, particularly the signaling lymphocyte activation molecule (SLAM) receptor binding site, correlate with adaptation to non-canine hosts. Differences in the phosphoprotein (P) gene sequences between disease and non-disease causing CDV strains may relate to pathogenicity in domestic dogs and wildlife. Of most concern are reports of CDV infection and disease in non-human primates raising the possibility of zoonosis. In this article we review the global occurrence of CDV and PDV, and present both historical and genetic information relating to these viruses crossing species barriers.


Subject(s)
Animals, Wild/virology , Distemper Virus, Canine/genetics , Distemper Virus, Phocine/genetics , Host Specificity , Morbillivirus Infections/veterinary , Morbillivirus/genetics , Animals , Cats , Cetacea/virology , Climate Change , Distemper Virus, Canine/pathogenicity , Distemper Virus, Phocine/pathogenicity , Dogs , Morbillivirus/pathogenicity , Morbillivirus/physiology , Pets/virology , Primates/virology , Viral Proteins/genetics
10.
Viruses ; 11(7)2019 07 03.
Article in English | MEDLINE | ID: mdl-31277275

ABSTRACT

Epidemiological reports of phocine distemper virus (PDV) and cetacean morbillivirus (CeMV) have accumulated since their discovery nearly 30 years ago. In this review, we focus on the interaction between these marine morbilliviruses and their major cellular receptor, the signaling lymphocyte activation molecule (SLAM). The three-dimensional crystal structure and homology models of SLAMs have demonstrated that 35 residues are important for binding to the morbillivirus hemagglutinin (H) protein and contribute to viral tropism. These 35 residues are essentially conserved among pinnipeds and highly conserved among the Caniformia, suggesting that PDV can infect these animals, but are less conserved among cetaceans. Because CeMV can infect various cetacean species, including toothed and baleen whales, the CeMV-H protein is postulated to have broader specificity to accommodate more divergent SLAM interfaces and may enable the virus to infect seals. In silico analysis of viral H protein and SLAM indicates that each residue of the H protein interacts with multiple residues of SLAM and vice versa. The integration of epidemiological, virological, structural, and computational studies should provide deeper insight into host specificity and switching of marine morbilliviruses.


Subject(s)
Morbillivirus Infections/veterinary , Morbillivirus Infections/virology , Morbillivirus/physiology , Seawater/virology , Signaling Lymphocytic Activation Molecule Family/metabolism , Animals , Caniformia/virology , Cetacea/virology , Distemper Virus, Phocine , Host Specificity , Lymphocyte Activation , Models, Molecular , Morbillivirus/classification , Morbillivirus/genetics , Morbillivirus Infections/epidemiology , Phylogeny , Protein Conformation , Signaling Lymphocytic Activation Molecule Family/chemistry , Signaling Lymphocytic Activation Molecule Family/genetics
11.
Can J Microbiol ; 65(11): 783-794, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31238018

ABSTRACT

SLAM (CD150) and nectin-4 are the major morbillivirus receptors responsible for virus pathogenesis and host range expansion. Recently, morbillivirus infections have been reported in unnatural hosts, including endangered species, posing a threat to their conservation. To understand the host range expansion of morbilliviruses, we generated the full-length sequences of morbillivirus receptors (goat, sheep, and dog SLAM, and goat nectin-4) and tried to correlate their role in determining host tropism. A high level of amino acid identity was observed between the sequences of related species, and phylogenetic reconstruction showed that the receptor sequences of carnivores, marine mammals, and small ruminants grouped separately. Analysis of the ligand binding region (V region; amino acid residues 52-136) of SLAM revealed high amino acid identity between small ruminants and bovine SLAMs. Comparison of canine SLAM with ruminants and non-canids SLAM revealed appreciable changes, including charge alterations. Significant differences between feline SLAM and canine SLAM have been reported. The binding motifs of nectin-4 genes (FPAG motif and amino acid residues 60, 62, and 63) were found to be conserved in sheep, goat, and dog. The differences reported in the binding region may be responsible for the level of susceptibility or resistance of a species to a particular morbillivirus.


Subject(s)
Mammals/genetics , Morbillivirus Infections/veterinary , Morbillivirus/physiology , Receptors, Virus/genetics , Amino Acid Sequence , Animals , Cats/genetics , Cattle/genetics , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Dogs/genetics , Goats/genetics , Host Specificity , Mammals/classification , Mammals/virology , Morbillivirus/genetics , Morbillivirus Infections/genetics , Morbillivirus Infections/metabolism , Morbillivirus Infections/virology , Phylogeny , Receptors, Virus/chemistry , Sequence Alignment , Sequence Analysis , Sheep/genetics , Signaling Lymphocytic Activation Molecule Family Member 1/chemistry , Signaling Lymphocytic Activation Molecule Family Member 1/genetics
12.
Viruses ; 11(2)2019 02 09.
Article in English | MEDLINE | ID: mdl-30744110

ABSTRACT

Paramyxoviruses comprise a large number of diverse viruses which in part give rise to severe diseases in affected hosts. A new genotype of feline morbillivirus, tentatively named feline morbillivirus genotype 2 (FeMV-GT2), was isolated from urine of cats with urinary tract diseases. Whole genome sequencing showed about 78% nucleotide homology to known feline morbilliviruses. The virus was isolated in permanent cell lines of feline and simian origin. To investigate the cell tropism of FeMV-GT2 feline primary epithelial cells from the kidney, the urinary bladder and the lung, peripheral blood mononuclear cells (PBMC), as well as organotypic brain slice cultures were used for infection experiments. We demonstrate that FeMV-GT2 is able to infect renal and pulmonary epithelial cells, primary cells from the cerebrum and cerebellum, as well as immune cells in the blood, especially CD4⁺ T cells, CD20⁺ B cells and monocytes. The cats used for virus isolation shed FeMV-GT2 continuously for several months despite the presence of neutralizing antibodies in the blood. Our results point towards the necessity of increased awareness for this virus when clinical signs of the aforementioned organs are encountered in cats which cannot be explained by other etiologies.


Subject(s)
Brain/virology , Kidney/virology , Leukocytes, Mononuclear/virology , Lung/virology , Morbillivirus Infections/veterinary , Morbillivirus/genetics , Animals , Brain/cytology , Cat Diseases/urine , Cat Diseases/virology , Cats , Cells, Cultured , Female , Genotype , Kidney/cytology , Lung/cytology , Male , Morbillivirus/physiology , Phylogeny , RNA, Viral , Viral Tropism , Whole Genome Sequencing
14.
Emerg Infect Dis ; 24(12): 2334-2337, 2018 12.
Article in English | MEDLINE | ID: mdl-30457523

ABSTRACT

Eradication of small ruminant morbillivirus (PPRV) is targeted for 2030. PPRV lineage IV is found in much of Asia and Africa. We used PPRV lineage IV strain Kurdistan/2011 in transmission trials to investigate the role of pigs, wild boar, and small ruminants as PPRV reservoirs. Suids were a possible source of infection.


Subject(s)
Animal Diseases/virology , Host Specificity , Morbillivirus/physiology , Ruminants/virology , Viral Tropism , Animal Diseases/diagnosis , Animal Diseases/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Goats , Morbillivirus/isolation & purification , Neutralization Tests , Swine
16.
Adv Virus Res ; 100: 75-98, 2018.
Article in English | MEDLINE | ID: mdl-29551144

ABSTRACT

Despite the availability of safe and effective vaccines against measles and several animal morbilliviruses, they continue to cause regular outbreaks and epidemics in susceptible populations. Morbilliviruses are highly contagious and share a similar pathogenesis in their respective hosts. This review provides an overview of morbillivirus history and the general replication cycle and recapitulates Morbillivirus pathogenesis focusing on common and unique aspects seen in different hosts. It also summarizes the state of knowledge regarding virus-host interactions on the cellular level with an emphasis on viral interference with innate immune response activation, and highlights remaining knowledge gaps.


Subject(s)
Host-Pathogen Interactions , Morbillivirus Infections/immunology , Morbillivirus Infections/virology , Morbillivirus/physiology , Animals , Humans , Immune Evasion , Morbillivirus/growth & development , Morbillivirus/immunology , Morbillivirus/pathogenicity , Virus Replication
17.
Curr Opin Virol ; 28: 133-141, 2018 02.
Article in English | MEDLINE | ID: mdl-29316493

ABSTRACT

Transmission of morbilliviruses within and among marine mammal species has been documented in a variety of marine habitats. Cetacean morbillivirus spreads between cetacean species in the aquatic environment whereas both phocine distemper virus and canine distemper virus have been associated with transmission within and between pinniped and terrestrial carnivore species in their natural habitat and at the aquatic-terrestrial interface. Periodically these viruses have caused large epizootics involving thousands of animals, due to sustained intra-species virus transmission. Social behavior of host species, marine habitat, geographical barriers and virus-host adaptations all likely contribute toward modulating virus spread. In combination with increased surveillance and whole genome sequencing, further research into ecological and host factors will be pivotal in better understanding the global transmission dynamics of marine morbilliviruses.


Subject(s)
Cetacea/virology , Morbillivirus Infections/transmission , Morbillivirus/physiology , Animals , Host Microbial Interactions , Morbillivirus/genetics , Oceans and Seas , Phylogeny
18.
Acta Virol ; 61(4): 495-497, 2017.
Article in English | MEDLINE | ID: mdl-29186969

ABSTRACT

Dolphin morbillivirus (DMV), a highly pathogenic agent, may cause peculiar, "brain-only" forms of infection (BOFDI), in which viral antigen and/or genome is found exclusively in the brain from striped dolphins (Stenella coeruleoalba). These BOFDIs show morphopathological similarities with subacute sclerosing panencephalitis and old dog encephalitis (ODE) in measles virus-infected patients and in canine distemper virus-infected dogs, respectively. The brain tissue from 3 BOFDI-affected striped dolphins was investigated by means of double labelling-indirect immunofluorescence (DL-IIF) and ultrastructurally, in order to characterize the DMV-targeted neuronal and non-neuronal cell populations, along with the associated submicroscopic findings. Viral colonization of calbindin-immunoreactive (IR) and nitric oxide synthase-IR neurons was detected in the cerebral parenchyma from the 3 DMV-infected dolphins under study, associated with nuclear (chromatin) and cytoplasmic (mitochondrial) ultrastructural changes. Furthermore, a limited viral targeting of brain astrocytes was found in these animals, all of which exhibited a prominent astrogliosis/astrocytosis. To the best of our knowledge, those herein reported should be the first submicroscopic pathology and neuropathogenetic data about BOFDI in striped dolphins. In this respect, the marked astrogliosis/astrocytosis and the low viral colonization of brain astrocytes in the 3 DMV-infected dolphins under investigation are of interest from the comparative pathology and viral neuropathogenesis standpoints, when compared with ODE-affected dogs, in whose brain a non-cytolytic, astrocyte-to-astrocyte infectious spread has been recently documented. Further studies aimed at characterizing the complex DMV-host interactions in BOFDI-affected striped dolphins are needed.


Subject(s)
Astrocytes/virology , Encephalitis/veterinary , Morbillivirus Infections/veterinary , Morbillivirus/physiology , Neurons/virology , Stenella/virology , Animals , Brain/cytology , Brain/virology , Encephalitis/virology , Morbillivirus/genetics , Morbillivirus Infections/virology
19.
PLoS Pathog ; 13(5): e1006371, 2017 May.
Article in English | MEDLINE | ID: mdl-28481926

ABSTRACT

Identification of cellular receptors and characterization of viral tropism in animal models have vastly improved our understanding of morbillivirus pathogenesis. However, specific aspects of viral entry, dissemination and transmission remain difficult to recapitulate in animal models. Here, we used three virologically identical but phenotypically distinct recombinant (r) canine distemper viruses (CDV) expressing different fluorescent reporter proteins for in vivo competition and airborne transmission studies in ferrets (Mustela putorius furo). Six donor ferrets simultaneously received three rCDVs expressing green, red or blue fluorescent proteins via conjunctival (ocular, Oc), intra-nasal (IN) or intra-tracheal (IT) inoculation. Two days post-inoculation sentinel ferrets were placed in physically separated adjacent cages to assess airborne transmission. All donor ferrets developed lymphopenia, fever and lethargy, showed progressively increasing systemic viral loads and were euthanized 14 to 16 days post-inoculation. Systemic replication of virus inoculated via the Oc, IN and IT routes was detected in 2/6, 5/6 and 6/6 ferrets, respectively. In five donor ferrets the IT delivered virus dominated, although replication of two or three different viruses was detected in 5/6 animals. Single lymphocytes expressing multiple fluorescent proteins were abundant in peripheral blood and lymphoid tissues, demonstrating the occurrence of double and triple virus infections. Transmission occurred efficiently and all recipient ferrets showed evidence of infection between 18 and 22 days post-inoculation of the donor ferrets. In all cases, airborne transmission resulted in replication of a single-colored virus, which was the dominant virus in the donor ferret. This study demonstrates that morbilliviruses can use multiple entry routes in parallel, and co-infection of cells during viral dissemination in the host is common. Airborne transmission was efficient, although transmission of viruses expressing a single color suggested a bottleneck event. The identity of the transmitted virus was not determined by the site of inoculation but by the viral dominance during dissemination.


Subject(s)
Distemper Virus, Canine/physiology , Ferrets , Morbillivirus Infections/virology , Morbillivirus/physiology , Animals , Chlorocebus aethiops , Coinfection , Genes, Reporter , Morbillivirus/pathogenicity , Morbillivirus Infections/transmission , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Vero Cells , Viral Load , Virus Internalization
20.
J Cell Mol Med ; 21(4): 816-830, 2017 04.
Article in English | MEDLINE | ID: mdl-27860224

ABSTRACT

Histiocytic sarcoma represents a rare malignant tumour with a short survival time, indicating the need of novel treatment strategies including oncolytic virotherapy. The underlying molecular mechanisms of viral oncolysis are largely unknown. As cancer in companion animals shares striking similarities with human counterparts, we chose a permanent canine histiocytic sarcoma cell line (DH82 cells) to identify global transcriptome changes following infection with canine distemper virus (CDV), a paramyxovirus closely related to human measles virus. Microarray analysis identified 3054 differentially expressed probe sets (DEPs), encoding for 892 up- and 869 down-regulated unique canine genes, respectively, in DH82 cells persistently infected with the vaccine strain Onderstepoort of CDV (DH82-Ond-pi), compared to non-infected DH82 cells. Up-regulated genes were predominantly related to immune processes, as demonstrated by functional enrichment analysis. Moreover, there was substantial enrichment of genes characteristic for classically activated M1 and alternatively activated M2 macrophages in DH82-Ond-pi; however, significant polarization into either of both categories was lacking. 'Angiogenesis' was the dominant enriched functional term for the down-regulated genes, highlighting decreased blood vessel generation as a potential mechanism of paramyxovirus-induced oncolysis in DH82 cells. The anti-angiogenic effect of infection was verified by immunohistochemistry, which revealed a lower blood vessel density in an in vivo mouse model, xenotransplanted with DH82-Ond-pi, compared to mice transplanted with non-infected DH82 cells. Reduction in angiogenesis appears to be an important oncolytic mechanism of CDV in DH82 cells, suggesting that similar mechanisms might account for human histiocytic sarcoma and maybe other tumours in conjunction with measles virus.


Subject(s)
Gene Expression Regulation, Neoplastic , Histiocytic Sarcoma/genetics , Histiocytic Sarcoma/therapy , Morbillivirus/physiology , Neovascularization, Pathologic/genetics , Oncolytic Virotherapy , Translational Research, Biomedical , Xenograft Model Antitumor Assays , Animals , Cell Line, Tumor , Cluster Analysis , Distemper Virus, Canine , Dogs , Down-Regulation/genetics , Gene Expression Profiling , Humans , Immunity/genetics , Macrophages/metabolism , Mice , Molecular Sequence Annotation , Necrosis , Neovascularization, Pathologic/pathology , Phenotype , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Principal Component Analysis , Remission Induction , Transcription, Genetic , Transcriptome/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...