Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Immunol ; 42(1): 551-584, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941604

ABSTRACT

Poxviruses have evolved a wide array of mechanisms to evade the immune response, and we provide an overview of the different immunomodulatory strategies. Poxviruses prevent the recognition of viral DNA that triggers the immune responses and inhibit signaling pathways within the infected cell. A unique feature of poxviruses is the production of secreted proteins that mimic cytokines and cytokine receptors, acting as decoy receptors to neutralize the activity of cytokines and chemokines. The capacity of these proteins to evade cellular immune responses by inhibiting cytokine activation is complemented by poxviruses' strategies to block natural killer cells and cytotoxic T cells, often through interfering with antigen presentation pathways. Mechanisms that target complement activation are also encoded by poxviruses. Virus-encoded proteins that target immune molecules and pathways play a major role in immune modulation, and their contribution to viral pathogenesis, facilitating virus replication or preventing immunopathology, is discussed.


Subject(s)
Immune Evasion , Poxviridae Infections , Poxviridae , Humans , Poxviridae/immunology , Poxviridae/physiology , Animals , Poxviridae Infections/immunology , Cytokines/metabolism , Signal Transduction , Viral Proteins/metabolism , Viral Proteins/immunology , Antigen Presentation/immunology , Host-Pathogen Interactions/immunology
2.
PLoS Pathog ; 17(8): e1009541, 2021 08.
Article in English | MEDLINE | ID: mdl-34437654

ABSTRACT

Herpes simplex virus type 1 and 2 (HSV-1 and HSV-2, respectively) are prevalent human pathogens of clinical relevance that establish long-life latency in the nervous system. They have been considered, along with the Herpesviridae family, to exhibit a low level of genetic diversity during viral replication. However, the high ability shown by these viruses to rapidly evolve under different selective pressures does not correlates with that presumed genetic stability. High-throughput sequencing has revealed that heterogeneous or plaque-purified populations of both serotypes contain a broad range of genetic diversity, in terms of number and frequency of minor genetic variants, both in vivo and in vitro. This is reminiscent of the quasispecies phenomenon traditionally associated with RNA viruses. Here, by plaque-purification of two selected viral clones of each viral subtype, we reduced the high level of genetic variability found in the original viral stocks, to more genetically homogeneous populations. After having deeply characterized the genetic diversity present in the purified viral clones as a high confidence baseline, we examined the generation of de novo genetic diversity under culture conditions. We found that both serotypes gradually increased the number of de novo minor variants, as well as their frequency, in two different cell types after just five and ten passages. Remarkably, HSV-2 populations displayed a much higher raise of nonconservative de novo minor variants than the HSV-1 counterparts. Most of these minor variants exhibited a very low frequency in the population, increasing their frequency over sequential passages. These new appeared minor variants largely impacted the coding diversity of HSV-2, and we found some genes more prone to harbor higher variability. These data show that herpesviruses generate de novo genetic diversity differentially under equal in vitro culture conditions. This might have contributed to the evolutionary divergence of HSV-1 and HSV-2 adapting to different anatomical niche, boosted by selective pressures found at each epithelial and neuronal tissue.


Subject(s)
Biological Evolution , Genetic Variation , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/genetics , Keratinocytes/virology , Virus Replication , Genome, Viral , Herpes Simplex/genetics , Herpes Simplex/metabolism , Humans , Keratinocytes/metabolism , Virus Activation , Virus Latency
3.
Proc Natl Acad Sci U S A ; 117(43): 26885-26894, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33046647

ABSTRACT

Ectromelia virus (ECTV) causes mousepox, a surrogate mouse model for smallpox caused by variola virus in humans. Both orthopoxviruses encode tumor necrosis factor receptor (TNFR) homologs or viral TNFR (vTNFR). These homologs are termed cytokine response modifier (Crm) proteins, containing a TNF-binding domain and a chemokine-binding domain called smallpox virus-encoded chemokine receptor (SECRET) domain. ECTV encodes one vTNFR known as CrmD. Infection of ECTV-resistant C57BL/6 mice with a CrmD deletion mutant virus resulted in uniform mortality due to excessive TNF secretion and dysregulated inflammatory cytokine production. CrmD dampened pathology, leukocyte recruitment, and inflammatory cytokine production in lungs including TNF, IL-6, IL-10, and IFN-γ. Blockade of TNF, IL-6, or IL-10R function with monoclonal antibodies reduced lung pathology and provided 60 to 100% protection from otherwise lethal infection. IFN-γ caused lung pathology only when both the TNF-binding and SECRET domains were absent. Presence of the SECRET domain alone induced significantly higher levels of IL-1ß, IL-6, and IL-10, likely overcoming any protective effects that might have been afforded by anti-IFN-γ treatment. The use of TNF-deficient mice and those that express only membrane-associated but not secreted TNF revealed that CrmD is critically dependent on host TNF for its function. In vitro, recombinant Crm proteins from different orthopoxviruses bound to membrane-associated TNF and dampened inflammatory gene expression through reverse signaling. CrmD does not affect virus replication; however, it provides the host advantage by enabling survival. Host survival would facilitate virus spread, which would also provide an advantage to the virus.


Subject(s)
Ectromelia virus/physiology , Host-Pathogen Interactions , Receptors, Tumor Necrosis Factor/metabolism , Respiratory Tract Infections/virology , Viral Proteins/metabolism , Animals , Cell Line , Chlorocebus aethiops , Female , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Respiratory Tract Infections/pathology , Viral Load
4.
J Gen Virol ; 103(10)2022 10.
Article in English | MEDLINE | ID: mdl-36260063

ABSTRACT

The application of CRISPR/Cas9 to improve genome engineering efficiency for large dsDNA viruses has been extensively described, but a robust and versatile method for high-throughput generation of marker-free recombinants for a desired locus has not yet been reported. Cytoplasmic-replicating viruses use their own repair enzymes for homologous recombination, while nuclear-replicating viruses use the host repair machinery. This is translated into a wide range of Cas9-induced homologous recombination efficiencies, depending on the virus replication compartment and viral/host repair machinery characteristics and accessibility. However, the use of Cas9 as a selection agent to target parental virus genomes robustly improves the selection of desired recombinants across large dsDNA viruses. We used ectromelia virus (ECTV) and herpes simplex virus (HSV) type 1 and 2 to optimize a CRISPR/Cas9 method that can be used versatilely for efficient genome editing and selection of both cytoplasmic- and nuclear-replicating viruses. We performed a genome-wide genetic variant analysis of mutations located at predicted off-target sequences for 20 different recombinants, showing off-target-free accuracy by deep sequencing. Our results support this optimized method as an efficient, accurate and versatile approach to enhance the two critical factors of high-throughput viral genome engineering: generation and colour-based selection of recombinants. This application of CRISPR/Cas9 reduces the time and labour for screening of desired recombinants, allowing for high-throughput generation of large collections of mutant dsDNA viruses for a desired locus, optimally in less than 2 weeks.


Subject(s)
Herpesvirus 1, Human , Viruses , CRISPR-Cas Systems , Gene Editing/methods , Genome, Viral , Herpesvirus 1, Human/genetics , Viruses/genetics
5.
J Virol ; 94(20)2020 09 29.
Article in English | MEDLINE | ID: mdl-32669337

ABSTRACT

During primary infection, herpes simplex virus 2 (HSV-2) replicates in epithelial cells and enters neurites to infect neurons of the peripheral nervous system. Growth factors and attractive and repulsive directional cues influence neurite outgrowth and neuronal survival. We hypothesized that HSV-2 modulates the activity of such cues to increase neurite outgrowth. To test this hypothesis, we exposed sensory neurons to nerve growth factor (NGF) and mock- or HSV-2-infected HEK-293T cells, since they express repellents of neurite outgrowth. We show that HEK-293T cells secrete factors that inhibit neurite outgrowth, while infection with HSV-2 strains MS and 333 reduces this repelling phenotype, increasing neurite numbers. The HSV-2-mediated restoration of neurite outgrowth required the activity of NGF. In the absence of infection, however, NGF did not overcome the repulsion mediated by HEK-293T cells. We previously showed that recombinant, soluble glycoprotein G of HSV-2 (rSgG2) binds and enhances NGF activity, increasing neurite outgrowth. However, the effect of gG2 during infection has not been investigated. Therefore, we addressed whether gG2 contributes to overcoming neurite outgrowth repulsion. To do so, we generated viruses lacking gG2 expression and complemented them by exogenous expression of gG2. Overall, our results suggest that HSV-2 infection of nonneuronal cells reduces their repelling effect on neurite outgrowth in an NGF-dependent manner. gG2 contributed to this phenotype, but it was not the only factor. The enhanced neurite outgrowth may facilitate HSV-2 spread from epithelial cells into neurons expressing NGF receptors and increase HSV-2-mediated pathogenesis.IMPORTANCE Herpes simplex virus 2 (HSV-2) is a prevalent human pathogen that establishes lifelong latency in neurons of the peripheral nervous system. Colonization of neurons is required for HSV-2 persistence and pathogenesis. The viral and cellular factors required for efficient infection of neurons are not fully understood. We show here that nonneuronal cells repel neurite outgrowth of sensory neurons, while HSV-2 infection overcomes this inhibition and, rather, stimulates neurite outgrowth. HSV-2 glycoprotein G and nerve growth factor contribute to this phenotype, which may attract neurites to sites of infection and facilitate virus spread to neurons. Understanding the mechanisms that modulate neurite outgrowth and facilitate HSV-2 infection of neurons might foster the development of therapeutics to reduce HSV-2 colonization of the nervous system and provide insights on neurite outgrowth and regeneration.


Subject(s)
Herpes Genitalis/metabolism , Herpesvirus 2, Human/metabolism , Nerve Growth Factor/metabolism , Neurites , Animals , Cell Line, Tumor , Chlorocebus aethiops , HEK293 Cells , Herpesvirus 2, Human/pathogenicity , Humans , Mice , Mice, Inbred BALB C , Neurites/metabolism , Neurites/virology , Vero Cells
6.
PLoS Pathog ; 15(4): e1007658, 2019 04.
Article in English | MEDLINE | ID: mdl-30947296

ABSTRACT

Throughout evolution, cytomegaloviruses (CMVs) have been capturing genes from their hosts, employing the derived proteins to evade host immune defenses. We have recently reported the presence of a number of CD48 homologs (vCD48s) encoded by different pathogenic viruses, including several CMVs. However, their properties and biological relevance remain as yet unexplored. CD48, a cosignaling molecule expressed on the surface of most hematopoietic cells, modulates the function of natural killer (NK) and other cytotoxic cells by binding to its natural ligand 2B4 (CD244). Here, we have characterized A43, the vCD48 exhibiting the highest amino acid sequence identity with host CD48. A43, which is encoded by owl monkey CMV, is a soluble molecule released from the cell after being proteolytically processed through its membrane proximal region. A43 is expressed with immediate-early kinetics, yielding a protein that is rapidly detected in the supernatant of infected cells. Remarkably, surface plasmon resonance assays revealed that this viral protein binds to host 2B4 with high affinity and slow dissociation rates. We demonstrate that soluble A43 is capable to abrogate host CD48:2B4 interactions. Moreover, A43 strongly binds to human 2B4 and prevents 2B4-mediated NK-cell adhesion to target cells, therefore reducing the formation of conjugates and the establishment of immunological synapses between human NK cells and CD48-expressing target cells. Furthermore, in the presence of this viral protein, 2B4-mediated cytotoxicity and IFN-γ production by NK cells are severely impaired. In summary, we propose that A43 may serve as a functional soluble CD48 decoy receptor by binding and masking 2B4, thereby impeding effective NK cell immune control during viral infections. Thus, our findings provide a novel example of the immune evasion strategies developed by viruses.


Subject(s)
CD48 Antigen/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus/immunology , Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Receptors, Immunologic/immunology , Signaling Lymphocytic Activation Molecule Family/immunology , CD48 Antigen/metabolism , Cells, Cultured , Cytomegalovirus Infections/virology , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/virology , Lymphocyte Activation , Receptors, Immunologic/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism
8.
J Biol Chem ; 294(13): 5214-5227, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30723161

ABSTRACT

Etanercept is a soluble form of the tumor necrosis factor receptor 2 (TNFR2) that inhibits pathological tumor necrosis factor (TNF) responses in rheumatoid arthritis and other inflammatory diseases. However, besides TNF, etanercept also blocks lymphotoxin-α (LTα), which has no clear therapeutic value and might aggravate some of the adverse effects associated with etanercept. Poxviruses encode soluble TNFR2 homologs, termed viral TNF decoy receptors (vTNFRs), that display unique specificity properties. For instance, cytokine response modifier D (CrmD) inhibits mouse and human TNF and mouse LTα, but it is inactive against human LTα. Here, we analyzed the molecular basis of these immunomodulatory activities in the ectromelia virus-encoded CrmD. We found that the overall molecular mechanism to bind TNF and LTα from mouse and human origin is fairly conserved in CrmD and dominated by a groove under its 50s loop. However, other ligand-specific binding determinants optimize CrmD for the inhibition of mouse ligands, especially mouse TNF. Moreover, we show that the inability of CrmD to inhibit human LTα is caused by a Glu-Phe-Glu motif in its 90s loop. Importantly, transfer of this motif to etanercept diminished its anti-LTα activity in >60-fold while weakening its TNF-inhibitory capacity in 3-fold. This new etanercept variant could potentially be used in the clinic as a safer alternative to conventional etanercept. This work is the most detailed study of the vTNFR-ligand interactions to date and illustrates that a better knowledge of vTNFRs can provide valuable information to improve current anti-TNF therapies.


Subject(s)
Ectromelia virus/immunology , Immunologic Factors/immunology , Lymphotoxin-alpha/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Tumor Necrosis Factor Decoy Receptors/immunology , Viral Proteins/immunology , Amino Acid Sequence , Animals , Ectromelia virus/chemistry , Ectromelia, Infectious/virology , Humans , Immunologic Factors/chemistry , Mice , Models, Molecular , Protein Domains , Tumor Necrosis Factor-alpha/immunology , Viral Proteins/chemistry
9.
J Biol Chem ; 293(45): 17418-17429, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30257868

ABSTRACT

Chemokines interact with glycosaminoglycans (GAGs) at the cellular surface and to specific cell-surface receptors to activate signaling pathways. The GAG interaction allows the formation of a chemotactic gradient of chemokine required for cell haptotaxis and chemokine oligomerization. Poxviruses encode secreted chemokine-binding proteins with no sequence similarity to their cellular counterparts to modulate the host immune system. The E163 protein from ectromelia virus, the causative agent of mousepox, binds chemokines through their GAG-binding domain. In addition, E163 interacts with GAGs to be anchored at the cell surface, but its ability to interfere with chemokine-GAG interactions has not been demonstrated. We report the identification of the GAG-binding regions in E163 and the generation of mutant forms deficient of GAG binding. Chemokine binding assays show that some of the E163 GAG-binding sites are also involved in the interaction with chemokines. By using recombinant GAG-binding mutant forms we demonstrate that E163 prevents the interaction of chemokines with cell-surface GAGs, providing mechanisms for the immunomodulatory activity of the viral chemokine-binding protein E163.


Subject(s)
Chemokines/chemistry , Ectromelia virus/chemistry , Glycosaminoglycans/chemistry , Viral Proteins/chemistry , Animals , CHO Cells , Chemokines/genetics , Chemokines/metabolism , Cricetulus , Ectromelia virus/genetics , Ectromelia virus/metabolism , Glycosaminoglycans/genetics , Glycosaminoglycans/metabolism , Mutation , Protein Binding , Protein Domains , Viral Proteins/genetics , Viral Proteins/metabolism
10.
EMBO J ; 33(6): 559-77, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24502978

ABSTRACT

T-cell receptors (TCR) recognize their antigen ligand at the interface between T cells and antigen-presenting cells, known as the immunological synapse (IS). The IS provides a means of sustaining the TCR signal which requires the continual supply of new TCRs. These are endocytosed and redirected from distal membrane locations to the IS. In our search for novel cytoplasmic effectors, we have identified ß-arrestin-1 as a ligand of non-phosphorylated resting TCRs. Using dominant-negative and knockdown approaches we demonstrate that ß-arrestin-1 is required for the internalization and downregulation of non-engaged bystander TCRs. Furthermore, TCR triggering provokes the ß-arrestin-1-mediated downregulation of the G-protein coupled chemokine receptor CXCR4, but not of other control receptors. We demonstrate that ß-arrestin-1 recruitment to the TCR, and bystander TCR and CXCR4 downregulation, are mechanistically mediated by the TCR-triggered PKC-mediated phosphorylation of ß-arrestin-1 at Ser163. This mechanism allows the first triggered TCRs to deliver a stop migration signal, and to promote the internalization of distal TCRs and CXCR4 and their translocation to the IS. This receptor crosstalk mechanism is critical to sustain the TCR signal.


Subject(s)
Arrestins/metabolism , Gene Expression Regulation/immunology , Immunological Synapses/metabolism , Models, Immunological , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Animals , Blotting, Western , Electroporation , Fluorescent Antibody Technique , Gene Knockdown Techniques , HEK293 Cells , Humans , Immunoprecipitation , Jurkat Cells , Mice , Mice, Transgenic , Microscopy, Fluorescence , Pyrimidines , Receptors, CXCR4/metabolism , Time-Lapse Imaging , beta-Arrestin 1 , beta-Arrestins
11.
J Virol ; 90(19): 8768-79, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27440877

ABSTRACT

UNLABELLED: Lymphocystis disease is a geographically widespread disease affecting more than 150 different species of marine and freshwater fish. The disease, provoked by the iridovirus lymphocystis disease virus (LCDV), is characterized by the appearance of papillomalike lesions on the skin of affected animals that usually self-resolve over time. Development of the disease is usually associated with several environmental factors and, more frequently, with stress conditions provoked by the intensive culture conditions present in fish farms. In gilthead sea bream (Sparus aurata), an economically important cultured fish species in the Mediterranean area, a distinct LCDV has been identified but not yet completely characterized. We have used direct sequencing of the virome of lymphocystis lesions from affected S. aurata fish to obtain the complete genome of a new LCDV-Sa species that is the largest vertebrate iridovirus sequenced to date. Importantly, this approach allowed us to assemble the full-length circular genome sequence of two previously unknown viruses belonging to the papillomaviruses and polyomaviruses, termed Sparus aurata papillomavirus 1 (SaPV1) and Sparus aurata polyomavirus 1 (SaPyV1), respectively. Epidemiological surveys showed that lymphocystis disease was frequently associated with the concurrent appearance of one or both of the new viruses. SaPV1 has unique characteristics, such as an intron within the L1 gene, and as the first member of the Papillomaviridae family described in fish, provides evidence for a more ancient origin of this family than previously thought. IMPORTANCE: Lymphocystis disease affects marine and freshwater fish species worldwide. It is characterized by the appearance of papillomalike lesions on the skin that contain heavily enlarged cells (lymphocysts). The causative agent is the lymphocystis disease virus (LCDV), a large icosahedral virus of the family Iridoviridae In the Mediterranean area, the gilthead sea bream (Sparus aurata), an important farmed fish, is frequently affected. Using next-generation sequencing, we have identified within S. aurata lymphocystis lesions the concurrent presence of an additional LCDV species (LCDV-Sa) as well as two novel viruses. These are members of polyomavirus and papillomavirus families, and here we report them to be frequently associated with the presence of lymphocysts in affected fish. Because papillomaviruses have not been described in fish before, these findings support a more ancient origin of this virus family than previously thought and evolutionary implications are discussed.


Subject(s)
Coinfection/veterinary , DNA Virus Infections/veterinary , Fish Diseases/virology , Iridoviridae/isolation & purification , Papillomaviridae/isolation & purification , Polyomavirus/isolation & purification , Sea Bream , Animals , Coinfection/pathology , Coinfection/virology , DNA Virus Infections/pathology , DNA Virus Infections/virology , DNA, Viral/chemistry , DNA, Viral/genetics , Fish Diseases/pathology , Iridoviridae/classification , Iridoviridae/genetics , Papillomaviridae/classification , Papillomaviridae/genetics , Polyomavirus/classification , Polyomavirus/genetics , Sequence Analysis, DNA
12.
PLoS Pathog ; 11(1): e1004571, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25611061

ABSTRACT

Herpes simplex virus type 1 (HSV-1) and HSV-2 are highly prevalent viruses that cause a variety of diseases, from cold sores to encephalitis. Both viruses establish latency in peripheral neurons but the molecular mechanisms facilitating the infection of neurons are not fully understood. Using surface plasmon resonance and crosslinking assays, we show that glycoprotein G (gG) from HSV-2, known to modulate immune mediators (chemokines), also interacts with neurotrophic factors, with high affinity. In our experimental model, HSV-2 secreted gG (SgG2) increases nerve growth factor (NGF)-dependent axonal growth of sympathetic neurons ex vivo, and modifies tropomyosin related kinase (Trk)A-mediated signaling. SgG2 alters TrkA recruitment to lipid rafts and decreases TrkA internalization. We could show, with microfluidic devices, that SgG2 reduced NGF-induced TrkA retrograde transport. In vivo, both HSV-2 infection and SgG2 expression in mouse hindpaw epidermis enhance axonal growth modifying the termination zone of the NGF-dependent peptidergic free nerve endings. This constitutes, to our knowledge, the discovery of the first viral protein that modulates neurotrophins, an activity that may facilitate HSV-2 infection of neurons. This dual function of the chemokine-binding protein SgG2 uncovers a novel strategy developed by HSV-2 to modulate factors from both the immune and nervous systems.


Subject(s)
Herpes Simplex/pathology , Nerve Endings/drug effects , Nerve Growth Factor/metabolism , Receptor, trkA/metabolism , Viral Envelope Proteins/pharmacology , Animals , Cells, Cultured , Chemotaxis/drug effects , HEK293 Cells , Herpes Simplex/metabolism , Herpesvirus 2, Human/metabolism , Herpesvirus 2, Human/pathogenicity , Humans , Mice , Nerve Endings/metabolism , Nerve Endings/pathology , Neurons/drug effects , Neurons/physiology , Signal Transduction/drug effects , Viral Envelope Proteins/metabolism
13.
Appl Environ Microbiol ; 83(13)2017 07 01.
Article in English | MEDLINE | ID: mdl-28455334

ABSTRACT

Pollen, fungi, and bacteria are the main microscopic biological entities present in outdoor air, causing allergy symptoms and disease transmission and having a significant role in atmosphere dynamics. Despite their relevance, a method for monitoring simultaneously these biological particles in metropolitan environments has not yet been developed. Here, we assessed the use of the Hirst-type spore trap to characterize the global airborne biota by high-throughput DNA sequencing, selecting regions of the 16S rRNA gene and internal transcribed spacer for the taxonomic assignment. We showed that aerobiological communities are well represented by this approach. The operational taxonomic units (OTUs) of two traps working synchronically compiled >87% of the total relative abundance for bacterial diversity collected in each sampler, >89% for fungi, and >97% for pollen. We found a good correspondence between traditional characterization by microscopy and genetic identification, obtaining more-accurate taxonomic assignments and detecting a greater diversity using the latter. We also demonstrated that DNA sequencing accurately detects differences in biodiversity between samples. We concluded that high-throughput DNA sequencing applied to aerobiological samples obtained with Hirst spore traps provides reliable results and can be easily implemented for monitoring prokaryotic and eukaryotic entities present in the air of urban areas.IMPORTANCE Detection, monitoring, and characterization of the wide diversity of biological entities present in the air are difficult tasks that require time and expertise in different disciplines. We have evaluated the use of the Hirst spore trap (an instrument broadly employed in aerobiological studies) to detect and identify these organisms by DNA-based analyses. Our results showed a consistent collection of DNA and a good concordance with traditional methods for identification, suggesting that these devices can be used as a tool for continuous monitoring of the airborne biodiversity, improving taxonomic resolution and characterization together. They are also suitable for acquiring novel DNA amplicon-based information in order to gain a better understanding of the biological particles present in a scarcely known environment such as the air.


Subject(s)
Air/analysis , Bacteria/isolation & purification , Eukaryota/isolation & purification , Fungi/isolation & purification , High-Throughput Nucleotide Sequencing/methods , Pollen/genetics , Air Microbiology , Bacteria/classification , Bacteria/genetics , Biodiversity , Cities , Eukaryota/classification , Eukaryota/genetics , Fungi/classification , Fungi/genetics , Phylogeny , Seasons , Spores, Fungal/classification , Spores, Fungal/genetics , Spores, Fungal/isolation & purification
14.
J Biol Chem ; 290(26): 15973-84, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25940088

ABSTRACT

The blockade of tumor necrosis factor (TNF) by etanercept, a soluble version of the human TNF receptor 2 (hTNFR2), is a well established strategy to inhibit adverse TNF-mediated inflammatory responses in the clinic. A similar strategy is employed by poxviruses, encoding four viral TNF decoy receptor homologues (vTNFRs) named cytokine response modifier B (CrmB), CrmC, CrmD, and CrmE. These vTNFRs are differentially expressed by poxviral species, suggesting distinct immunomodulatory properties. Whereas the human variola virus and mouse ectromelia virus encode one vTNFR, the broad host range cowpox virus encodes all vTNFRs. We report the first comprehensive study of the functional and binding properties of these four vTNFRs, providing an explanation for their expression profile among different poxviruses. In addition, the vTNFRs activities were compared with the hTNFR2 used in the clinic. Interestingly, CrmB from variola virus, the causative agent of smallpox, is the most potent TNFR of those tested here including hTNFR2. Furthermore, we demonstrate a new immunomodulatory activity of vTNFRs, showing that CrmB and CrmD also inhibit the activity of lymphotoxin ß. Similarly, we report for the first time that the hTNFR2 blocks the biological activity of lymphotoxin ß. The characterization of vTNFRs optimized during virus-host evolution to modulate the host immune response provides relevant information about their potential role in pathogenesis and may be used to improve anti-inflammatory therapies based on soluble decoy TNFRs.


Subject(s)
Cowpox virus/metabolism , Poxviridae/metabolism , Receptors, Tumor Necrosis Factor, Type II/chemistry , Receptors, Tumor Necrosis Factor, Type II/metabolism , Tumor Necrosis Factor Decoy Receptors/chemistry , Tumor Necrosis Factor Decoy Receptors/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cowpox virus/chemistry , Cowpox virus/genetics , Humans , Lymphotoxin-beta/metabolism , Mice , Molecular Sequence Data , Poxviridae/chemistry , Poxviridae/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Sequence Alignment , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factors/metabolism , Viral Proteins/genetics
15.
J Gen Virol ; 97(11): 3007-3016, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27692045

ABSTRACT

Herpes simplex virus type 1 and type 2 (HSV-1 and HSV-2, respectively) are among the most prevalent human pathogens, causing a variety of diseases. HSV modulation of the chemokine network remains poorly understood. We have previously identified secreted glycoprotein G (SgG) as the first viral chemokine-binding protein that enhances chemokine function as a novel viral immunomodulatory mechanism. However, gG is also present at the viral envelope and its role in the virus particle remains unknown. Here we have addressed the chemokine-binding capacity of HSV particles and the functionality of such interaction in vitro. We adapted surface plasmon resonance assays and demonstrated the ability of HSV particles to bind a specific set of human chemokines with high affinity. Moreover, we identified gG as the envelope glycoprotein mediating such interaction, as shown by the lack of binding to a HSV-1 gG mutant. In contrast to HSV-1, HSV-2 gG is cleaved and the chemokine-binding domain is secreted (SgG2). However, we found that HSV-2 particles retain the ability to bind chemokines, potentially through SgG2 associated to the viral envelope or non-processed precursor protein. Moreover, we found that HSV particles increase cell migration independently of chemokine binding to envelope gG. This work provides insights into HSV manipulation of the host immune system.


Subject(s)
Cell Movement , Chemokines/metabolism , Herpes Simplex/physiopathology , Herpesvirus 1, Human/metabolism , Herpesvirus 2, Human/metabolism , Viral Envelope Proteins/metabolism , Chemokines/genetics , Herpes Simplex/genetics , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 2, Human/genetics , Host-Pathogen Interactions , Humans , Protein Binding , Viral Envelope Proteins/genetics
16.
J Neuroinflammation ; 13(1): 210, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27576911

ABSTRACT

Genital herpes is a painful disease frequently caused by the neurotropic pathogen herpes simplex virus type 2 (HSV-2). We have recently shown that HSV-2-secreted glycoprotein G (SgG2) interacts with and modulates the activity of the neurotrophin nerve growth factor (NGF). This interaction modifies the response of the NGF receptor TrkA, increasing NGF-dependent axonal growth. NGF is not only an axonal growth modulator but also an important mediator of pain and inflammation regulating the amount, localization, and activation of the thermal pain receptor transient receptor potential vanilloid 1 (TRPV1). In this work, we addressed whether SgG2 could contribute to HSV-2-induced pain. Injection of SgG2 in the mouse hindpaw produced a rapid and transient increase in thermal pain sensitivity. At the molecular level, this acute increase in thermal pain induced by SgG2 injection was dependent on differential NGF-induced phosphorylation and in changes in the amount of TrkA and TRPV1 in the dermis. These results suggest that SgG2 alters thermal pain sensitivity by modulating TRPV1 receptor.


Subject(s)
Nerve Growth Factor/toxicity , Pain Threshold/physiology , Pain/chemically induced , Pain/metabolism , TRPV Cation Channels/metabolism , Viral Envelope Proteins/toxicity , Animals , Animals, Newborn , Cells, Cultured , Hot Temperature/adverse effects , Male , Mice , Pain Threshold/drug effects
17.
J Virol ; 89(20): 10702-6, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26246565

ABSTRACT

Amphibian-like ranaviruses include pathogens of fish, amphibians, and reptiles that have recently evolved from a fish-infecting ancestor. The molecular determinants of host range and virulence in this group are largely unknown, and currently fish infection models are lacking. We show that European sheatfish virus (ESV) can productively infect zebrafish, causing a lethal pathology, and describe a method for the generation of recombinant ESV, establishing a useful model for the study of fish ranavirus infections.


Subject(s)
DNA Virus Infections/veterinary , Disease Models, Animal , Fish Diseases/virology , Ranavirus/genetics , Zebrafish/virology , Animals , Base Sequence , DNA Virus Infections/pathology , DNA Virus Infections/virology , Fish Diseases/pathology , Genetic Engineering , Genotype , Larva/virology , Molecular Sequence Data , Phylogeny , Ranavirus/classification , Ranavirus/pathogenicity , Virulence
18.
J Gen Virol ; 96(10): 3118-3123, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26242179

ABSTRACT

Poxviruses encode up to four different soluble TNF receptors, named cytokine response modifier B (CrmB), CrmC, CrmD and CrmE. These proteins mimic the extracellular domain of the cellular TNF receptors to bind and inhibit the activity of TNF and, in some cases, other TNF superfamily ligands. Most of these ligands are released after the enzymic cleavage of a membrane precursor. However, transmembrane TNF (tmTNF) is not only a precursor of soluble TNF but also exerts specific pro-inflammatory and immunological activities. Here, we report that viral TNF receptors bound and inhibited tmTNF and describe some interesting differences in their activity against the soluble cytokine. Thus, CrmE, which does not inhibit mouse soluble TNF, could block murine tmTNF-induced cytotoxicity. We propose that this anti-tmTNF effect should be taken into consideration when assessing the role of viral TNF decoy receptors in the pathogenesis of poxvirus.


Subject(s)
Immune Evasion , Poxviridae/immunology , Poxviridae/physiology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Viral Proteins/metabolism , Animals , Cell Line , Fibroblasts/immunology , Fibroblasts/virology , Macrophages/immunology , Macrophages/virology , Mice
19.
Nat Rev Immunol ; 3(1): 36-50, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12511874

ABSTRACT

Viruses have evolved elegant mechanisms to evade detection and destruction by the host immune system. One of the evasion strategies that have been adopted by large DNA viruses is to encode homologues of cytokines, chemokines and their receptors--molecules that have a crucial role in control of the immune response. Viruses have captured host genes or evolved genes to target specific immune pathways, and so viral genomes can be regarded as repositories of important information about immune processes, offering us a viral view of the host immune system. The study of viral immunomodulatory proteins might help us to uncover new human genes that control immunity, and their characterization will increase our understanding of not only viral pathogenesis, but also normal immune mechanisms. Moreover, viral proteins indicate strategies of immune modulation that might have therapeutic potential.


Subject(s)
Chemokines/immunology , DNA Viruses/immunology , Molecular Mimicry/immunology , Receptors, Chemokine/immunology , Receptors, Cytokine/immunology , Viral Proteins/immunology , Chemokines/metabolism , Cytokines/immunology , Cytokines/metabolism , DNA Virus Infections/immunology , DNA Virus Infections/metabolism , DNA Viruses/metabolism , Humans , Models, Molecular , Receptors, Chemokine/metabolism , Receptors, Cytokine/metabolism , Viral Proteins/metabolism
20.
BMC Genomics ; 15: 989, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25407630

ABSTRACT

BACKGROUND: Viruses have unique properties, small genome and regions of high similarity, whose effects on metagenomic assemblies have not been characterized so far. This study uses diverse in silico simulated viromes to evaluate how extensively genomes can be assembled using different sequencing platforms and assemblers. Further, it investigates the suitability of different methods to estimate viral diversity in metagenomes. RESULTS: We created in silico metagenomes mimicking various platforms at different sequencing depths. The CLC assembler revealed subpar compared to IDBA_UD and CAMERA , which are metagenomic-specific. Up to a saturation point, Illumina platforms proved more capable of reconstructing large portions of viral genomes compared to 454. Read length was an important factor for limiting chimericity, while scaffolding marginally improved contig length and accuracy. The genome length of the various viruses in the metagenomes did not significantly affect genome reconstruction, but the co-existence of highly similar genomes was detrimental. When evaluating diversity estimation tools, we found that PHACCS results were more accurate than those from CatchAll and clustering, which were both orders of magnitude above expected. CONCLUSIONS: Assemblers designed specifically for the analysis of metagenomes should be used to facilitate the creation of high-quality long contigs. Despite the high coverage possible, scientists should not expect to always obtain complete genomes, because their reconstruction may be hindered by co-existing species bearing highly similar genomic regions. Further development of metagenomics-oriented assemblers may help bypass these limitations in future studies. Meanwhile, the lack of fully reconstructed communities keeps methods to estimate viral diversity relevant. While none of the three methods tested had absolute precision, only PHACCS was deemed suitable for comparative studies.


Subject(s)
Genetic Variation , Genome, Viral , Metagenome/genetics , Metagenomics/methods , Contig Mapping
SELECTION OF CITATIONS
SEARCH DETAIL