Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Immunity ; 57(5): 1056-1070.e5, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38614091

ABSTRACT

A specialized population of mast cells residing within epithelial layers, currently known as intraepithelial mast cells (IEMCs), was originally observed over a century ago, yet their physiological functions have remained enigmatic. In this study, we unveil an unexpected and crucial role of IEMCs in driving gasdermin C-mediated type 2 immunity. During helminth infection, αEß7 integrin-positive IEMCs engaged in extensive intercellular crosstalk with neighboring intestinal epithelial cells (IECs). Through the action of IEMC-derived proteases, gasdermin C proteins intrinsic to the epithelial cells underwent cleavage, leading to the release of a critical type 2 cytokine, interleukin-33 (IL-33). Notably, mast cell deficiency abolished the gasdermin C-mediated immune cascade initiated by epithelium. These findings shed light on the functions of IEMCs, uncover a previously unrecognized phase of type 2 immunity involving mast cell-epithelial cell crosstalk, and advance our understanding of the cellular mechanisms underlying gasdermin C activation.


Subject(s)
Interleukin-33 , Mast Cells , Phosphate-Binding Proteins , Mast Cells/immunology , Mast Cells/metabolism , Animals , Interleukin-33/metabolism , Interleukin-33/immunology , Mice , Phosphate-Binding Proteins/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Cell Communication/immunology
2.
Rheumatology (Oxford) ; 63(3): 856-865, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37467058

ABSTRACT

OBJECTIVE: Accumulating evidence from microbial studies have highlighted the modulatory roles of intestinal microbes in numerous human diseases, however, the shared microbial signatures across different diseases remain relatively unclear. METHODS: To consolidate existing knowledge across multiple studies, we performed meta-analyses of 17 disease types, covering 34 case-control datasets of 16S rRNA sequencing data, to identify shared alterations among different diseases. Furthermore, the impact of a microbial species, Lactobacillus salivarius, was established in a dextran sodium sulphate-induced colitis model and a collagen type II-induced arthritis mouse model. RESULTS: Microbial alterations among autoimmune diseases were substantially more consistent compared with that of other diseases (cancer, metabolic disease and nervous system disease), with microbial signatures exhibiting notable discriminative power for disease prediction. Autoimmune diseases were characterized by the enrichment of Enterococcus, Veillonella, Streptococcus and Lactobacillus and the depletion of Ruminococcus, Gemmiger, Oscillibacter, Faecalibacterium, Lachnospiracea incertae sedis, Anaerostipes, Coprococcus, Alistipes, Roseburia, Bilophila, Barnesiella, Dorea, Ruminococcus2, Butyricicoccus, Phascolarctobacterium, Parabacteroides and Odoribacter, among others. Functional investigation of L. salivarius, whose genus was commonly enriched in numerous autoimmune diseases, demonstrated protective roles in two separate inflammatory mouse models. CONCLUSION: Our study highlights a strong link between autoimmune diseases and the gut microbiota, with notably consistent microbial alterations compared with that of other diseases, indicating that therapeutic strategies that target the gut microbiome may be transferable across different autoimmune diseases. Functional validation of L. salivarius highlighted that bacterial genera associated with disease may not always be antagonistic, but may represent protective or adaptive responses to disease.


Subject(s)
Arthritis, Experimental , Autoimmune Diseases , Gastrointestinal Microbiome , Animals , Mice , Humans , RNA, Ribosomal, 16S , Clostridiales , Disease Models, Animal
3.
Cell Rep ; 42(7): 112754, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37405914

ABSTRACT

As highly organized consortia of bacteria, biofilms have long been implicated in aggravating inflammation. However, our understanding regarding in vivo host-biofilm interactions in the complex tissue environments remains limited. Here, we show a unique pattern of crypt occupation by mucus-associated biofilms during the early stage of colitis, which is genetically dependent on bacterial biofilm-forming capacity and restricted by host epithelial α1,2-fucosylation. α1,2-Fucosylation deficiency leads to markedly augmented crypt occupation by biofilms originated from pathogenic Salmonella Typhimurium or indigenous Escherichia coli, resulting in exacerbated intestinal inflammation. Mechanistically, α1,2-fucosylation-mediated restriction of biofilms relies on interactions between bacteria and liberated fucose from biofilm-occupied mucus. Fucose represses biofilm formation and biofilm-related genes in vitro and in vivo. Finally, fucose administration ameliorates experimental colitis, suggesting therapeutic potential of fucose for biofilm-related disorders. This work illustrates host-biofilm interactions during gut inflammation and identifies fucosylation as a physiological strategy for restraining biofilm formation.


Subject(s)
Colitis , Fucose , Humans , Biofilms , Colitis/microbiology , Glycosylation , Bacteria , Inflammation
4.
Mucosal Immunol ; 15(2): 279-288, 2022 02.
Article in English | MEDLINE | ID: mdl-34697434

ABSTRACT

Well-orchestrated transcriptional programs in intestinal epithelial cells (IECs) are essential for maintenance of optimal mucosal barrier functions, whereas the contribution of elongation-related mechanisms to barrier function remains unknown. Here, a combination of genetic and genomic approaches defined a critical role of IEC-intrinsic negative elongation factor (NELF) complex in maintenance of epithelial homeostasis. By direct occupancy at endogenous gene loci, NELF sustained expression of a subset of genes related to junctional integrity. As a result, epithelial NELF deficiency results in subdued levels of these junction-related genes and excessive IEC necroptosis in vivo secondary to commensal microbial invasion. In a colitis model, NELF-deficient mice exhibited severely impaired barrier integrity characterized by increased intestinal permeability and significantly exacerbated intestinal inflammation with lethal consequences. Our findings reveal the protective function of the NELF complex against intestinal damage and inflammation and suggest that elongation represents a biologically important step in defining IEC transcriptome.


Subject(s)
Colitis , Intestinal Mucosa , Transcription Factors , Animals , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Permeability , Transcription Factors/metabolism
5.
Front Microbiol ; 12: 728823, 2021.
Article in English | MEDLINE | ID: mdl-34721326

ABSTRACT

Gut microbiome dysbiosis has been known to be associated with all stages of non-alcoholic fatty liver disease (NAFLD), but questions remain about microbial profiles in progression and homogeneity across NAFLD stages. We performed a meta-analysis of three publicly shotgun datasets and built predictive models to determine diagnostic capacity. Here, we found consistently microbiome shifts across NAFLD stages, of which co-occurrence patterns and core sets of new biomarkers significantly correlated with NAFLD progression were identified. Machine learning models that are able to distinguish patients with any NAFLD stage from healthy controls remained predictive when applied to patients with other NAFLD stages, suggesting the homogeneity across stages once again. Focusing on species and metabolic pathways specifically associated with progressive stages, we found that increased toxic metabolites and decreased protection of butyrate and choline contributed to advanced NAFLD. We further built models discriminating one stage from the others with an average of 0.86 of area under the curve. In conclusion, this meta-analysis firmly establishes generalizable microbiome dysbiosis and predictive taxonomic and functional signatures as a basis for future diagnostics across NAFLD stages.

6.
Front Immunol ; 11: 2065, 2020.
Article in English | MEDLINE | ID: mdl-33013873

ABSTRACT

In addition to their established functions in host defense, accumulating evidence has suggested an emerging role for antimicrobial proteins (AMPs) in shaping commensal microbiota. However, the role of α-defensins, the most abundant AMPs of intestine, in regulating microbial ecology remains inconclusive. Here, we report that α-defensins promote commensal Bacteroides colonization by enhancing bacterial adhesion to the mucosal reservoir. Experiments utilizing mice deficient in matrix metalloproteinase 7 (MMP7), the α-defensin-activating enzyme, with rigorous littermate controls showed that α-defensin deficiency did not significantly influence steady-state intestinal microbiota. In contrast, α-defensins are essential for replenishment of commensal Bacteroides from the mucosal reservoir following antibiotics-induced dysbiosis, shown by markedly compromised recovery of Bacteroides in Mmp7-/- animals. Mechanistically, α-defensins promote Bacteroides colonization on epithelial surfaces in vivo and adhesion to epithelial cells in vitro. Moreover, α-defensins unexpectedly does not show any microbicidal activities against Bacteroides. Together, we propose that α-defensins promote commensal bacterial colonization and recovery to maintain microbial diversity upon environmental challenges.


Subject(s)
Anti-Bacterial Agents/adverse effects , Bacteroides Infections/immunology , Bacteroides/physiology , Drug-Related Side Effects and Adverse Reactions/immunology , Dysbiosis/immunology , Intestinal Mucosa/immunology , alpha-Defensins/metabolism , Animals , Anti-Bacterial Agents/therapeutic use , Homeostasis , Matrix Metalloproteinase 7/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Ribosomal, 16S/genetics , Symbiosis
7.
Cell Host Microbe ; 25(5): 706-718.e7, 2019 05 08.
Article in English | MEDLINE | ID: mdl-31053533

ABSTRACT

Metabolic programs and host defense are highly integrated to ensure proper immune responses during stress. Central to these responses, mTOR regulates immune functions by sensing and integrating environmental cues, yet how these systems are coordinated at the intestinal surface remains undefined. We show that the antimicrobial peptide α-defensin is functionally sustained during nutrient deprivation because of regulation of the defensin-processing enzyme MMP7 by microbiota- and host-derived factors. Unlike other antimicrobial peptides, the MMP7-α-defensin axis remains active during nutrient fluctuations, providing essential protection against enteric pathogens. Sustained Mmp7 expression requires the microbiota and is mediated by de-repression of the transcription activator Atoh1 upon attenuation of the transcriptional repressor Hes1 in intestinal epithelial cells. Hes1 levels are regulated via mTOR and controlled translationally, constituting a metabolism-translation-transcription loop. Disrupting this loop by supplying nutrients paradoxically compromises antibacterial defense. Together, these results uncover a regulatory circuit that couples host nutrient status to epithelial antimicrobial immunity.


Subject(s)
Epithelial Cells/immunology , Gene Expression Regulation , Immunity, Mucosal , Matrix Metalloproteinase 7/biosynthesis , Nutrients/metabolism , Transcription Factor HES-1/metabolism , alpha-Defensins/biosynthesis , Animals , Cell Line , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Mice, Inbred C57BL
8.
Virology ; 485: 135-44, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26247624

ABSTRACT

There is an urgent need to develop new vaccines against highly pathogenic PRRS virus (HP-PRRSV) variant in China. The actual use of each codon pairs is more or less frequent than that of the statistical prediction and codon pair bias (CPB) usage affects gene translation. We "shuffled" the existing codons in HP-PRRSV genes GP5, M, nsp2 and nsp9, so that the CPB of these genes could be more negative. De-optimization of nsp9, the RNA-dependent RNA polymerase, significantly decreased PRRSV replication in porcine alveolar macrophages (PAMs). In vitro study showed that HV-nsp9(min) and HV-nsp29(min) were remarkably attenuated in PAMs, and inoculation of pigs with 2 ml⁎10(5.0) TCID50/ml of HV-nsp9(min) or HV-nsp29(min) did not cause PRRS. Importantly, pigs immunized with HV-nsp29(min) were fully protected against different HP-PRRSV strains׳ lethal challenges. Our results imply that the CPB de-optimized HV-nsp29(min) has the potential to be used as a live vaccine candidate against HP-PRRSV.


Subject(s)
Codon/chemistry , Macrophages, Alveolar/drug effects , Porcine Reproductive and Respiratory Syndrome/prevention & control , Porcine respiratory and reproductive syndrome virus/drug effects , RNA-Dependent RNA Polymerase/immunology , Viral Proteins/immunology , Viral Vaccines/immunology , Animals , Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Codon/immunology , Genetic Engineering , Immunization , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine Reproductive and Respiratory Syndrome/mortality , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/immunology , Protein Biosynthesis , RNA-Dependent RNA Polymerase/administration & dosage , RNA-Dependent RNA Polymerase/genetics , Survival Analysis , Swine , Vaccines, Attenuated , Viral Load , Viral Proteins/administration & dosage , Viral Proteins/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Virus Replication/drug effects
9.
Virology ; 468-470: 96-103, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25155198

ABSTRACT

Protein kinase C (PKC) that transduces signals to modulate a wide range of cellular functions has been shown to regulate a number of viral infections. Herein, we showed that inhibition of PKC with the PKC inhibitor GF109203X significantly impaired porcine reproductive and respiratory syndrome virus (PRRSV) replication. Inhibition of PKC led to virus yield reduction, which was associated with decreased viral RNA synthesis and lowered virus protein expression. And this inhibitory effect by PKC inhibitor was shown to occur at the early stage of PRRSV infection. Subsequently, we found that PRRSV infection activated PKCδ in PAMs and knockdown of PKCδ by small interfering RNA (siRNA) suppressed PRRSV replication, suggesting that novel PKCδ may play an important factor in PRRSV replication. Taken together, these data imply that PKC is involved in PRRSV infection and beneficial to PRRSV replication, extending our understanding of PRRSV replication.


Subject(s)
Gene Expression Regulation, Viral/physiology , Porcine respiratory and reproductive syndrome virus/physiology , Protein Kinase C-delta/metabolism , Virus Replication/physiology , Animals , Cells, Cultured , Gene Expression Regulation, Enzymologic , Indoles/pharmacology , Macrophages, Alveolar/virology , Maleimides/pharmacology , Protein Kinase C-delta/antagonists & inhibitors , RNA, Viral/genetics , RNA, Viral/metabolism , Swine , Viral Proteins/genetics , Viral Proteins/metabolism
10.
Vaccine ; 32(43): 5740-8, 2014 Sep 29.
Article in English | MEDLINE | ID: mdl-25171845

ABSTRACT

Transcription regulatory sequences (TRSs) play a key role in the synthesis of porcine reproductive and respiratory syndrome virus (PRRSV) subgenomic mRNAs, which resembles similarity-assisted RNA recombination. In this study, genome instability was found when a highly pathogenic PRRSV (HP-PRRSV) strain was inserted by an additional transcription unit in which a foreign gene GFP was expressed from TRS2 while a copy of TRS6 drove ORF2a/b transcription. Structural protein gene-deleted genomes resulted from enhanced RNA recombinations were identified in the recombinant virus rHV-GFP. Moreover, rHV-GFP replicated slower than parental viruses, and caused less cell death in porcine alveolar macrophages. Pigs infected with rHV-GFP survived with no or mild syndromes, whereas all pigs infected with parental viruses died within 12 days. Our data showed that additional transcription unit insertion could confer genome instability and attenuation of HP-PRRSV.


Subject(s)
Genome, Viral , Genomic Instability , Mutagenesis, Insertional , Porcine respiratory and reproductive syndrome virus/genetics , Animals , Base Sequence , Gene Deletion , Molecular Sequence Annotation , Nucleic Acid Conformation , Porcine Reproductive and Respiratory Syndrome/immunology , RNA, Viral/genetics , Sus scrofa , Swine
11.
J Virol ; 88(18): 10934-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25008936

ABSTRACT

UNLABELLED: Porcine reproductive and respiratory syndrome virus (PRRSV) is a highly infectious pathogen that causes severe diseases in pigs and great economic losses to the swine industry worldwide. Type I interferons (IFNs) play a crucial role in antiviral immunity. In the present study, we demonstrated that infection with the highly pathogenic PRRSV strain JXwn06 antagonized type I IFN expression induced by poly(I·C) in both porcine alveolar macrophages (PAMs) and blood monocyte-derived macrophages (BMo). Subsequently, we showed that the inhibition of poly(I·C)-induced IFN-ß production by PRRSV was dependent on the blocking of NF-κB signaling pathways. By screening PRRSV nonstructural and structural proteins, we demonstrated that nonstructural protein 4 (nsp4), a viral 3C-like serine protease, significantly suppressed IFN-ß expression. Moreover, we verified that nsp4 inhibited NF-κB activation induced by signaling molecules, including RIG-I, VISA, TRIF, and IKKß. nsp4 was shown to target the NF-κB essential modulator (NEMO) at the E349-S350 site to mediate its cleavage. Importantly, nsp4 mutants with defective protease activity abolished its ability to cleave NEMO and inhibit IFN-ß production. These findings might have implications for our understanding of PRRSV pathogenesis and its mechanisms for evading the host immune response. IMPORTANCE: Porcine reproductive and respiratory syndrome virus (PRRSV) is a major agent of respiratory diseases in pigs. Like many other viruses, PRRSV has evolved a variety of strategies to evade host antiviral innate immunity for survival and propagation. In this study, we show that PRRSV nsp4 is a novel antagonist of the NF-κB signaling pathway, which is responsible for regulating the expression of type I interferons and other crucial cytokines. We then investigated the underlying mechanism used by nsp4 to suppress NF-κB-mediated IFN-ß production. We found that nsp4 interfered with the NF-κB signaling pathway through the cleavage of NEMO (a key regulator of NF-κB signaling) at the E349-S350 site, leading to the downregulation of IFN-ß production induced by poly(I·C). The data presented here may help us to better understand PRRSV pathogenesis.


Subject(s)
Interferon-beta/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine respiratory and reproductive syndrome virus/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Down-Regulation , Host-Pathogen Interactions , Interferon-beta/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/metabolism , Macrophages/virology , NF-kappa B/genetics , NF-kappa B/metabolism , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , Signal Transduction , Swine , Viral Nonstructural Proteins/genetics
12.
Vet Microbiol ; 170(1-2): 48-57, 2014 May 14.
Article in English | MEDLINE | ID: mdl-24581811

ABSTRACT

Atypical porcine reproductive and respiratory syndrome (PRRS) caused by a highly pathogenic PRRS virus (HP-PRRSV) is characterized by high fever, high morbidity, high mortality, and associated with severe neurological symptoms. Microglia are the resident innate immune cells in central nervous system (CNS), and their activation has been implicated as an important contributor to the pathogenesis of CNS diseases. In the present study, we successfully cultured porcine microglia and demonstrated that microglia could support PRRSV infection and replication in vitro. We further showed that HP-PRRSV infection significantly up-regulated the key inflammatory factors including IL-1ß, TNF-α, IL-6, IL-12, IL-8, CXCL10, MCP-1, CCL3, CCL4, and CCL5 in cultured microglia as well as in the CNS of HP-PRRSV-infected pigs. The transcription factors NF-κB and AP-1, which are widely reported to regulate cytokine and chemokine productions, were activated by HP-PRRSV infection in microglia. Meanwhile, we found that HP-PRRSV induced cellular ROS formation in microglia and ROS scavenger was proved to significantly abolish the activation of pro-inflammatory cytokines (IL-1ß, TNF-α, IL-6, and IL-8), suggesting that ROS are crucial for pro-inflammatory gene production. Importantly, incubation with supernatants from HP-PRRSV-infected microglia cell culture remarkably induced SH-SY5Y neuroblastoma cell death. Collectively, these results showed that PRRSV infection induced cytokine and ROS up-regulation in microglia, which might contribute to neurotoxicity. These data have implications for us to understand the neuropathogenesis of HP-PRRSV in pigs.


Subject(s)
Cytokines/genetics , Microglia/immunology , Microglia/virology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/immunology , Up-Regulation , Animals , Brain/immunology , Brain/physiopathology , Cells, Cultured , Culture Media, Conditioned/toxicity , Cytokines/metabolism , Neurons/drug effects , Nitrous Oxide/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Swine
13.
Virology ; 450-451: 182-95, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24503081

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression post-transcriptionally and play critical roles in intricate networks of host-pathogen interactions and innate immunity. Porcine reproductive and respiratory syndrome (PRRS) is one of the most important diseases affecting swine industry worldwide. Here, we demonstrated that miR-23, miR-378, and miR-505 were antiviral host factors against PRRS virus (PRRSV). Over-expression of the three miRNAs inhibited PRRSV infection in a dose-dependent manner, respectively. Blockage of the three endogenously expressed miRNAs significantly enhanced PRRSV replication. Different type 2 PRRSV strains harbored conserved miR-23, miR-378, and miR-505 target sites (TSs) that were sufficient to confer miRNA-mediated repression of PRRSV replication. Interestingly, miR-23 was capable of inducing type I interferon expression during PRRSV infection through IRF3/IRF7 activation, which might further lead to the inhibition of virus infection. These results suggest that miR-23, miR-378, and miR-505, especially miR-23, may have the potential to be used as antiviral therapy against PRRSV infection.


Subject(s)
Interferon Type I/genetics , MicroRNAs/genetics , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine respiratory and reproductive syndrome virus/physiology , RNA, Viral/genetics , Up-Regulation , Virus Replication , Animals , Interferon Type I/metabolism , MicroRNAs/metabolism , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine Reproductive and Respiratory Syndrome/virology , Porcine respiratory and reproductive syndrome virus/genetics , RNA Interference , RNA, Viral/metabolism , Swine
14.
J Virol ; 88(5): 2810-20, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24352469

ABSTRACT

UNLABELLED: Atypical porcine reproductive and respiratory syndrome (PRRS) caused by highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) is characterized by high fever and high mortality. However, the mechanism underlying the fever induction is still unknown. Prostaglandin E2 (PGE2), synthesized by cyclooxygenase type 1/2 (COX-1/2) enzymes, is essential for inducing fever. In this study, we found that PGE2, together with COX-1, was significantly elevated by HP-PRRSV. We subsequently demonstrated that extracellular signal-regulated kinase 1/2 (ERK1/2) and phosphorylated ERK (p-ERK) were the key nodes to trigger COX-1 expression after HP-PRRSV infection. Furthermore, we proved the direct binding of p-C/EBP-ß to the COX-1 promoter by luciferase reporter and chromatin immunoprecipitation assays. In addition, silencing of C/EBP-ß remarkably impaired the enhancement of COX-1 production induced by HP-PRRSV infection. Taken together, our results indicate that HP-PPRSV elicits the expression of COX-1 through the ERK1/2-p-C/EBP-ß signaling pathway, resulting in the increase of PGE2, which might be the cause of high fever in infected pigs. Our findings might provide new insights into the molecular mechanisms underlying the pathogenesis of HP-PRRSV infection. IMPORTANCE: The atypical PRRS caused by HP-PRRSV was characterized by high fever, high morbidity, and high mortality in pigs of all ages, yet how HP-PRRSV induces high fever in pigs remains unknown. In the present study, we found out that HP-PRRSV infection could increase PGE2 production by upregulation of COX-1, and we subsequently characterized the underlying mechanisms about how HP-PRRSV enhances COX-1 production. PGE2 plays a critical role in inducing high temperature in hosts during pathogen infections. Thus, our findings here could help us have a better understanding of HP-PRRSV pathogenesis.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta/metabolism , Cyclooxygenase 1/metabolism , Dinoprostone/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Porcine respiratory and reproductive syndrome virus/physiology , Signal Transduction , Animals , Base Sequence , Cloning, Molecular , Cyclooxygenase 1/genetics , Molecular Sequence Data , Phosphorylation , Porcine Reproductive and Respiratory Syndrome/genetics , Porcine Reproductive and Respiratory Syndrome/metabolism , Porcine respiratory and reproductive syndrome virus/pathogenicity , Promoter Regions, Genetic , Response Elements , Swine
15.
J Virol ; 87(15): 8808-12, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23740977

ABSTRACT

We previously showed that microRNA 181 (miR-181) can inhibit PRRSV replication by directly targeting its genomic RNA. Here, we report that miR-181 can downregulate the PRRSV receptor CD163 in blood monocytes and porcine alveolar macrophages (PAMs) through targeting the 3' untranslated region (UTR) of CD163 mRNA. Downregulation of CD163 leads to the inhibition of PRRSV entry into PAMs and subsequently suppresses PRRSV infection. Our findings indicate that delivery of miR-181 can be used as antiviral therapy against PRRSV infection.


Subject(s)
MicroRNAs/metabolism , Porcine respiratory and reproductive syndrome virus/growth & development , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Virus/antagonists & inhibitors , Virus Internalization , Animals , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Macrophages, Alveolar/virology , MicroRNAs/genetics , Monocytes/virology , Porcine respiratory and reproductive syndrome virus/physiology , Receptors, Cell Surface/genetics , Receptors, Virus/genetics , Swine
16.
J Virol ; 87(2): 1159-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23152505

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important viral pathogens in the swine industry. Emerging evidence indicates that the host microRNAs (miRNAs) are involved in host-pathogen interactions. However, whether host miRNAs can target PRRSV and be used to inhibit PRRSV infection has not been reported. Recently, microRNA 181 (miR-181) has been identified as a positive regulator of immune response, and here we report that miR-181 can directly impair PRRSV infection. Our results showed that delivered miR-181 mimics can strongly inhibit PRRSV replication in vitro through specifically binding to a highly (over 96%) conserved region in the downstream of open reading frame 4 (ORF4) of the viral genomic RNA. The inhibition of PRRSV replication was specific and dose dependent. In PRRSV-infected Marc-145 cells, the viral mRNAs could compete with miR-181-targeted sequence in luciferase vector to interact with miR-181 and result in less inhibition of luciferase activity, further demonstrating the specific interactions between miR-181 and PRRSV RNAs. As expected, miR-181 and other potential PRRSV-targeting miRNAs (such as miR-206) are expressed much more abundantly in minimally permissive cells or tissues than in highly permissive cells or tissues. Importantly, highly pathogenic PRRSV (HP-PRRSV) strain-infected pigs treated with miR-181 mimics showed substantially decreased viral loads in blood and relief from PRRSV-induced fever compared to negative-control (NC)-treated controls. These results indicate the important role of host miRNAs in modulating PRRSV infection and viral pathogenesis and also support the idea that host miRNAs could be useful for RNA interference (RNAi)-mediated antiviral therapeutic strategies.


Subject(s)
MicroRNAs/metabolism , Porcine respiratory and reproductive syndrome virus/physiology , RNA, Viral/metabolism , Virus Replication , Animals , Cell Line , MicroRNAs/genetics , MicroRNAs/immunology , Porcine respiratory and reproductive syndrome virus/genetics , Porcine respiratory and reproductive syndrome virus/immunology , RNA, Viral/genetics , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...