Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Microbiol Resour Announc ; 10(21): e0033421, 2021 May 27.
Article in English | MEDLINE | ID: mdl-34042483

ABSTRACT

Dermatophilus congolensis causes dermatophilosis in cattle, mainly in tropical climates. Despite the economic losses caused by this bacterium, its pathogenic factors are less well understood. We report draft genomes of D. congolensis strains isolated during a dermatophilosis outbreak in cattle in St. Kitts and Nevis. Some isolates contain tet(Z), which is responsible for resistance to tetracyclines.

2.
Front Cell Neurosci ; 14: 544612, 2020.
Article in English | MEDLINE | ID: mdl-33281554

ABSTRACT

TLR3 provides immediate type I IFN response following entry of stimulatory PAMPs into the CNS, as it is in HSV infection. The receptor plays a vital role in astrocytes, contributing to rapid infection sensing and suppression of viral replication, precluding the spread of virus beyond neurons. The route of TLR3 mobilization culminating in the receptor activation remains unexplained. In this research, we investigated the involvement of various types of endosomes in the regulation of the TLR3 mobility in C8-D1A murine astrocyte cell line. TLR3 was transported rapidly to early EEA1-positive endosomes as well as LAMP1-lysosomes following stimulation with the poly(I:C). Later, TLR3 largely associated with late Rab7-positive endosomes. Twenty-four hours after stimulation, TLR3 co-localized with LAMP1 abundantly in lysosomes of astrocytes. TLR3 interacted with poly(I:C) intracellularly from 1 min to 8 h following cell stimulation. We detected TLR3 on the surface of astrocytes indicating constitutive expression, which increased after poly(I:C) stimulation. Our findings contribute to the understanding of cellular modulation of TLR3 trafficking. Detailed analysis of the TLR3 transportation pathway is an important component in disclosing the fate of the receptor in HSV-infected CNS and may help in the search for rationale therapeutics to control the replication of neuropathic viruses.

3.
Pathogens ; 9(10)2020 Oct 04.
Article in English | MEDLINE | ID: mdl-33020446

ABSTRACT

Dendritic cells (DCs) and macrophages are the first line of antiviral immunity. Viral pathogens exploit these cell populations for their efficient replication and dissemination via the modulation of intracellular signaling pathways. Disruption of the noncanonical nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) signaling has frequently been observed in lymphoid cells upon infection with oncogenic viruses. However, several nononcogenic viruses have been shown to manipulate the noncanonical NF-κB signaling in different cell types. This study demonstrates the modulating effect of ectromelia virus (ECTV) on the components of the noncanonical NF-κB signaling pathway in established murine cell lines: JAWS II DCs and RAW 264.7 macrophages. ECTV affected the activation of TRAF2, cIAP1, RelB, and p100 upon cell treatment with both canonical and noncanonical NF-κB stimuli and thus impeded DNA binding by RelB and p52. ECTV also inhibited the expression of numerous genes related to the noncanonical NF-κB pathway and RelB-dependent gene expression in the cells treated with canonical and noncanonical NF-κB activators. Thus, our data strongly suggest that ECTV influenced the noncanonical NF-κB signaling components in the in vitro models. These findings provide new insights into the noncanonical NF-κB signaling components and their manipulation by poxviruses in vitro.

4.
Immunol Invest ; 49(3): 232-248, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31240969

ABSTRACT

Ectromelia virus (ECTV), an orthopoxvirus, undergoes productive replication in conventional dendritic cells (cDCs), resulting in the inhibition of their innate and adaptive immune functions. ECTV replication rate in cDCs is increased due to downregulation of the expression of cathepsins - cystein proteases that orchestrate several steps during DC maturation. Therefore, this study was aimed to determine if downregulation of cathepsins, such as B, L or S, disrupts cDC capacity to induce activating signals in T cells or whether infection of cDCs with ECTV further weakens their functions as antigen-presenting cells. Our results showed that cDCs treated with siRNA against cathepsin B, L and S synthesize similar amounts of pro-inflammatory cytokines and exhibit comparable ability to mature and stimulate alloreactive CD4+ T cells, as untreated wild type (WT) cells. Moreover, ECTV inhibitory effect on cDC innate and adaptive immune functions, observed especially after LPS treatment, was comparable in both cathepsin-silenced and WT cells. Taken together, the absence of cathepsins B, L and S has minimal, if any, impact on the inhibitory effect of ECTV on cDC immune functions. We assume that the virus-mediated inhibition of cathepsin expression in cDCs represents more a survival mechanism than an immune evasion strategy.


Subject(s)
Cathepsins/deficiency , Dendritic Cells/immunology , Ectromelia virus/physiology , Animals , CD4-Positive T-Lymphocytes/immunology , Cathepsins/genetics , Cathepsins/metabolism , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/virology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Th1-Th2 Balance
5.
Arch Immunol Ther Exp (Warsz) ; 67(6): 401-414, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31324924

ABSTRACT

Poxviruses utilize multiple strategies to prevent activation of extrinsic and intrinsic apoptotic pathways for successful replication. Mitochondrial heat shock proteins (mtHsps), especially Hsp60 and its cofactor Hsp10, are engaged in apoptosis regulation; however, until now, the influence of poxviruses on mtHsps has never been studied. We used highly infectious Moscow strain of ectromelia virus (ECTV) to investigate the mitochondrial heat shock response and apoptotic potential in permissive L929 fibroblasts. Our results show that ECTV-infected cells exhibit mostly mitochondrial localization of Hsp60 and Hsp10, and show overexpression of both proteins during later stages of infection. ECTV infection has only moderate effect on the electron transport chain subunit expression. Moreover, increase of mtHsp amounts is accompanied by lack of apoptosis, and confirmed by reduced level of pro-apoptotic Bax protein and elevated levels of anti-apoptotic Bcl-2 and Bcl-xL proteins. Taken together, we show a positive relationship between increased levels of Hsp60 and Hsp10 and decreased apoptotic potential of L929 fibroblasts, and further hypothesize that Hsp60 and/or its cofactor play important roles in maintaining protein homeostasis in mitochondria for promotion of cell survival allowing efficient replication of ECTV.


Subject(s)
Chaperonin 10/metabolism , Chaperonin 60/metabolism , Ectromelia virus/physiology , Ectromelia, Infectious/immunology , Fibroblasts/physiology , Heat-Shock Response/immunology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Apoptosis , Cell Line , Fibroblasts/virology , Gene Expression Regulation , Immune Evasion , Mice , Protein Transport , Proto-Oncogene Proteins c-bcl-2/metabolism , Virulence , Virus Replication
6.
BMC Microbiol ; 19(1): 92, 2019 05 10.
Article in English | MEDLINE | ID: mdl-31077130

ABSTRACT

BACKGROUND: Cathepsins are a group of endosomal proteases present in many cells including dendritic cells (DCs). The activity of cathepsins is regulated by their endogenous inhibitors - cystatins. Cathepsins are crucial to antigen processing during viral and bacterial infections, and as such are a prerequisite to antigen presentation in the context of major histocompatibility complex class I and II molecules. Due to the involvement of DCs in both innate and adaptive immune responses, and the quest to understand the impact of poxvirus infection on host cells, we investigated the influence of ectromelia virus (ECTV) infection on cathepsin and cystatin levels in murine conventional DCs (cDCs). ECTV is a poxvirus that has evolved many mechanisms to avoid host immune response and is able to replicate productively in DCs. RESULTS: Our results showed that ECTV-infection of JAWS II DCs and primary murine GM-CSF-derived bone marrow cells down-regulated both mRNA and protein of cathepsin B, L and S, and cystatin B and C, particularly during the later stages of infection. Moreover, the activity of cathepsin B, L and S was confirmed to be diminished especially at later stages of infection in JAWS II cells. Consequently, ECTV-infected DCs had diminished ability to endocytose and process a soluble antigen. Close examination of cellular protein distribution showed that beginning from early stages of infection, the remnants of cathepsin L and cystatin B co-localized and partially co-localized with viral replication centers (viral factories), respectively. Moreover, viral yield increased in cDCs treated with siRNA against cathepsin B, L or S and subsequently infected with ECTV. CONCLUSIONS: Taken together, our results indicate that infection of cDCs with ECTV suppresses cathepsins and cystatins, and alters their cellular distribution which impairs the cDC function. We propose this as an additional viral strategy to escape immune responses, enabling the virus to replicate effectively in infected cells.


Subject(s)
Cathepsins/genetics , Cystatins/genetics , Dendritic Cells/virology , Ectromelia virus/physiology , Animals , Dendritic Cells/immunology , Down-Regulation , Endosomes/immunology , Endosomes/virology , Gene Knockdown Techniques , Male , Mice , Mice, Inbred C57BL , RNA, Small Interfering , Virus Replication
7.
Oxid Med Cell Longev ; 2019: 6927380, 2019.
Article in English | MEDLINE | ID: mdl-31089414

ABSTRACT

Toll-like receptors (TLRs) sense the presence of pathogen-associated molecular patterns. Nevertheless, the mechanisms modulating TLR-triggered innate immune responses are not yet fully understood. Complex regulatory systems exist to appropriately direct immune responses against foreign or self-nucleic acids, and a critical role of hepatocyte growth factor-regulated tyrosine kinase substrate (HRS), endosomal sorting complex required for transportation-0 (ESCRT-0) subunit, has recently been implicated in the endolysosomal transportation of TLR7 and TLR9. We investigated the involvement of Syk, Hrs, and STAM in the regulation of the TLR3 signaling pathway in a murine astrocyte cell line C8-D1A following cell stimulation with a viral dsRNA mimetic. Our data uncover a relationship between TLR3 and ESCRT-0, point out Syk as dsRNA-activated kinase, and suggest the role for Syk in mediating TLR3 signaling in murine astrocytes. We show molecular events that occur shortly after dsRNA stimulation of astrocytes and result in Syk Tyr-342 phosphorylation. Further, TLR3 undergoes proteolytic processing; the resulting TLR3 N-terminal form interacts with Hrs. The knockdown of Syk and Hrs enhances TLR3-mediated antiviral response in the form of IFN-ß, IL-6, and CXCL8 secretion. Understanding the role of Syk and Hrs in TLR3 immune responses is of high importance since activation and precise execution of the TLR3 signaling pathway in the brain seem to be particularly significant in mounting an effective antiviral defense. Infection of the brain with herpes simplex type 1 virus may increase the secretion of amyloid-ß by neurons and astrocytes and be a causal factor in degenerative diseases such as Alzheimer's disease. Errors in TLR3 signaling, especially related to the precise regulation of the receptor transportation and degradation, need careful observation as they may disclose foundations to identify novel or sustain known therapeutic targets.


Subject(s)
Antiviral Agents/metabolism , Astrocytes/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Phosphoproteins/metabolism , Syk Kinase/metabolism , Toll-Like Receptor 3/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Ligands , Mice , NF-kappa B/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Poly I-C/pharmacology , Protein Binding/drug effects , Signal Transduction/drug effects , Toll-Like Receptor 3/chemistry , Up-Regulation/drug effects
8.
Viruses ; 10(5)2018 05 16.
Article in English | MEDLINE | ID: mdl-29772718

ABSTRACT

Mitochondria are multifunctional organelles that participate in numerous processes in response to viral infection, but they are also a target for viruses. The aim of this study was to define subcellular events leading to alterations in mitochondrial morphology and function during infection with ectromelia virus (ECTV). We used two different cell lines and a combination of immunofluorescence techniques, confocal and electron microscopy, and flow cytometry to address subcellular changes following infection. Early in infection of L929 fibroblasts and RAW 264.7 macrophages, mitochondria gathered around viral factories. Later, the mitochondrial network became fragmented, forming punctate mitochondria that co-localized with the progeny virions. ECTV-co-localized mitochondria associated with the cytoskeleton components. Mitochondrial membrane potential, mitochondrial fission⁻fusion, mitochondrial mass, and generation of reactive oxygen species (ROS) were severely altered later in ECTV infection leading to damage of mitochondria. These results suggest an important role of mitochondria in supplying energy for virus replication and morphogenesis. Presumably, mitochondria participate in transport of viral particles inside and outside of the cell and/or they are a source of membranes for viral envelope formation. We speculate that the observed changes in the mitochondrial network organization and physiology in ECTV-infected cells provide suitable conditions for viral replication and morphogenesis.


Subject(s)
Ectromelia virus/physiology , Fibroblasts/metabolism , Fibroblasts/virology , Macrophages/metabolism , Macrophages/virology , Mitochondria/physiology , Mitochondria/ultrastructure , Animals , Autophagy/physiology , Dynamins/metabolism , Ectromelia virus/ultrastructure , Fibroblasts/pathology , GTP Phosphohydrolases/metabolism , L Cells , Macrophages/pathology , Membrane Potential, Mitochondrial/physiology , Mice , Microtubule-Organizing Center/metabolism , Microtubule-Organizing Center/virology , Mitochondria/metabolism , Mitochondria/virology , Mitochondrial Proteins/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/analysis , Tubulin/metabolism , Virion/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...