Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Proc Natl Acad Sci U S A ; 119(32): e2204539119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35878012

ABSTRACT

Viruses evade the innate immune response by suppressing the production or activity of cytokines such as type I interferons (IFNs). Here we report the discovery of a mechanism by which the SARS-CoV-2 virus coopts an intrinsic cellular machinery to suppress the production of the key immunostimulatory cytokine IFN-ß. We reveal that the SARS-CoV-2 encoded nonstructural protein 2 (NSP2) directly interacts with the cellular GIGYF2 protein. This interaction enhances the binding of GIGYF2 to the mRNA cap-binding protein 4EHP, thereby repressing the translation of the Ifnb1 mRNA. Depletion of GIGYF2 or 4EHP significantly enhances IFN-ß production, which inhibits SARS-CoV-2 replication. Our findings reveal a target for rescuing the antiviral innate immune response to SARS-CoV-2 and other RNA viruses.


Subject(s)
COVID-19 , Carrier Proteins , Interferon Type I , Viral Nonstructural Proteins , COVID-19/genetics , Carrier Proteins/metabolism , Cell Line , Eukaryotic Initiation Factor-4E/metabolism , Humans , Immunity, Innate , Interferon Type I/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , SARS-CoV-2 , Viral Nonstructural Proteins/metabolism , Virus Replication
2.
Nat Chem Biol ; 18(1): 81-90, 2022 01.
Article in English | MEDLINE | ID: mdl-34754101

ABSTRACT

Emerging evidence suggests that host glycans influence severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Here, we reveal that the receptor-binding domain (RBD) of the spike (S) protein on SARS-CoV-2 recognizes oligosaccharides containing sialic acid (Sia), with preference for monosialylated gangliosides. Gangliosides embedded within an artificial membrane also bind to the RBD. The monomeric affinities (Kd = 100-200 µM) of gangliosides for the RBD are similar to another negatively charged glycan ligand of the RBD proposed as a viral co-receptor, heparan sulfate (HS) dp2-dp6 oligosaccharides. RBD binding and infection of SARS-CoV-2 pseudotyped lentivirus to angiotensin-converting enzyme 2 (ACE2)-expressing cells is decreased following depletion of cell surface Sia levels using three approaches: sialyltransferase (ST) inhibition, genetic knockout of Sia biosynthesis, or neuraminidase treatment. These effects on RBD binding and both pseudotyped and authentic SARS-CoV-2 viral entry are recapitulated with pharmacological or genetic disruption of glycolipid biosynthesis. Together, these results suggest that sialylated glycans, specifically glycolipids, facilitate viral entry of SARS-CoV-2.


Subject(s)
Glycolipids/metabolism , SARS-CoV-2/metabolism , Sialic Acids/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Binding Sites , Humans
3.
Proc Natl Acad Sci U S A ; 117(43): 26926-26935, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33046650

ABSTRACT

Influenza virus infections cause a wide variety of outcomes, from mild disease to 3 to 5 million cases of severe illness and ∼290,000 to 645,000 deaths annually worldwide. The molecular mechanisms underlying these disparate outcomes are currently unknown. Glycosylation within the human host plays a critical role in influenza virus biology. However, the impact these modifications have on the severity of influenza disease has not been examined. Herein, we profile the glycomic host responses to influenza virus infection as a function of disease severity using a ferret model and our lectin microarray technology. We identify the glycan epitope high mannose as a marker of influenza virus-induced pathogenesis and severity of disease outcome. Induction of high mannose is dependent upon the unfolded protein response (UPR) pathway, a pathway previously shown to associate with lung damage and severity of influenza virus infection. Also, the mannan-binding lectin (MBL2), an innate immune lectin that negatively impacts influenza outcomes, recognizes influenza virus-infected cells in a high mannose-dependent manner. Together, our data argue that the high mannose motif is an infection-associated molecular pattern on host cells that may guide immune responses leading to the concomitant damage associated with severity.


Subject(s)
Glycoproteins/metabolism , Host-Pathogen Interactions , Influenza, Human/metabolism , Lung/metabolism , Mannose/metabolism , A549 Cells , Animals , Carbohydrate Metabolism , Female , Ferrets , Glycomics , Glycosylation , Humans , Influenza A Virus, H1N1 Subtype , Mannose-Binding Lectin/metabolism , X-Box Binding Protein 1/metabolism
4.
J Virol ; 95(13): e0026621, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34110264

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic. While previous studies have shown that several SARS-CoV-2 proteins can antagonize the interferon (IFN) response, some of the mechanisms by which they do so are not well understood. In this study, we describe two novel mechanisms by which SARS-CoV-2 blocks the IFN pathway. Type I IFNs and IFN-stimulated genes (ISGs) were poorly induced during SARS-CoV-2 infection, and once infection was established, cells were highly resistant to ectopic induction of IFNs and ISGs. Levels of two key IFN signaling pathway components, Tyk2 and STAT2, were significantly lower in SARS-CoV-2-infected cells. Expression of nonstructural protein 1 (NSP1) or nucleocapsid in the absence of other viral proteins was sufficient to block IFN induction, but only NSP1 was able to inhibit IFN signaling. Mapping studies suggest that NSP1 prevents IFN induction in part by blocking IRF3 phosphorylation. In addition, NSP1-induced depletion of Tyk2 and STAT2 dampened ISG induction. Together, our data provide new insights into how SARS-CoV-2 successfully evades the IFN system to establish infection. IMPORTANCE SARS-CoV-2 is the causative agent of COVID-19, a serious disease that can have a myriad of symptoms from loss of taste and smell to pneumonia and hypercoagulation. The rapid spread of SARS-CoV-2 can be attributed in part to asymptomatic transmission, where infected individuals shed large amounts of virus before the onset of disease. This is likely due to the ability of SARS-CoV-2 to effectively suppress the innate immune system, including the IFN response. Indeed, we show that the IFN response is efficiently blocked during SARS-CoV-2 infection, a process that is mediated in large part by nonstructural protein 1 and nucleocapsid. Our study provides new insights on how SARS-CoV-2 evades the IFN response to successfully establish infection. These findings should be considered for the development and administration of therapeutics against SARS-CoV-2.


Subject(s)
Interferon Type I/antagonists & inhibitors , SARS-CoV-2/metabolism , Signal Transduction , Viral Nonstructural Proteins/metabolism , Animals , COVID-19/immunology , COVID-19/virology , Chlorocebus aethiops , Coronavirus Nucleocapsid Proteins/metabolism , HEK293 Cells , Humans , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Phosphoproteins/metabolism , SARS-CoV-2/pathogenicity , STAT2 Transcription Factor/metabolism , TYK2 Kinase/metabolism , Vero Cells
5.
Appl Environ Microbiol ; 86(12)2020 06 02.
Article in English | MEDLINE | ID: mdl-32276973

ABSTRACT

Communities of gut bacteria (microbiota) are known to play roles in resistance to pathogen infection and optimal weight gain in turkey flocks. However, knowledge of turkey respiratory microbiota and its link to gut microbiota is lacking. This study presents a 16S rRNA gene-based census of the turkey respiratory microbiota (nasal cavity and trachea) alongside gut microbiota (cecum and ileum) in two identical commercial Hybrid Converter turkey flocks raised in parallel under typical field commercial conditions. The flocks were housed in adjacent barns during the brood stage and in geographically separated farms during the grow-out stage. Several bacterial taxa, primarily Staphylococcus, that were acquired in the respiratory tract at the beginning of the brood stage persisted throughout the flock cycle. Late-emerging predominant taxa in the respiratory tract included Deinococcus and Corynebacterium Tracheal and nasal microbiota of turkeys were identifiably distinct from one another and from gut microbiota. Nevertheless, gut and respiratory microbiota changed in parallel over time and appeared to share many taxa. During the brood stage, the two flocks generally acquired similar gut and respiratory microbiota, and their average body weights were comparable. However, there were qualitative and quantitative differences in microbial profiles and body weight gain trajectories after the flocks were transferred to geographically separated grow-out farms. Lower weight gain corresponded to the emergence of Deinococcus and Ornithobacterium in the respiratory tract and Fusobacterium and Parasutterella in gut. This study provides an overview of turkey microbiota under field conditions and suggests several hypotheses concerning the respiratory microbiome.IMPORTANCE Turkey meat is an important source of animal protein, and the industry around its production contributes significantly to the agricultural economy. The microorganisms present in the gut of turkeys are known to impact bird health and flock performance. However, the respiratory microbiota in turkeys is entirely unexplored. This study has elucidated the microbiota of respiratory tracts of turkeys from two commercial flocks raised in parallel throughout a normal flock cycle. Further, the study suggests that bacteria originating in the gut or in poultry house environments influence respiratory communities; consequently, they induce poor performance, either directly or indirectly. Future attempts to develop microbiome-based interventions for turkey health should delimit the contributions of respiratory microbiota and aim to limit disturbances to those communities.


Subject(s)
Cecum/microbiology , Ileum/microbiology , Microbiota , Nasal Cavity/microbiology , Trachea/microbiology , Turkeys/microbiology , Weight Gain , Animals , Bacterial Physiological Phenomena , Body-Weight Trajectory , Gastrointestinal Microbiome , Male
6.
Appl Environ Microbiol ; 85(9)2019 05 01.
Article in English | MEDLINE | ID: mdl-30824436

ABSTRACT

The digestive and respiratory tracts of chickens are colonized by bacteria that are believed to play important roles in the overall health and performance of the birds. Most of the current research on the commensal bacteria (microbiota) of chickens has focused on broilers and gut microbiota, and less attention has been given to layers and respiratory microbiota. This research bias has left significant gaps in our knowledge of the layer microbiome. This study was conducted to define the core microbiota colonizing the upper respiratory tract (URT) and lower intestinal tract (LIT) in commercial layers under field conditions. One hundred eighty-one chickens were sampled from a flock of >80,000 birds at nine times to collect samples for 16S rRNA gene-based bacterial metabarcoding. Generally, the body site and age/farm stage had very dominant effects on the quantity, taxonomic composition, and dynamics of core bacteria. Remarkably, ileal and URT microbiota were compositionally more related to each other than to that from the cecum. Unique taxa dominated in each body site yet some taxa overlapped between URT and LIT sites, demonstrating a common core. The overlapping bacteria also contained various levels of several genera with well-recognized avian pathogens. Our findings suggest that significant interaction exists between gut and respiratory microbiota, including potential pathogens, in all stages of the farm sequence. The baseline data generated in this study can be useful for the development of effective microbiome-based interventions to enhance production performance and to prevent and control disease in commercial chicken layers.IMPORTANCE The poultry industry is faced with numerous challenges associated with infectious diseases and suboptimal performance of flocks. As microbiome research continues to grow, it is becoming clear that poultry health and production performance are partly influenced by nonpathogenic symbionts that occupy different habitats within the bird. This study has defined the baseline composition and overlaps between respiratory and gut bacteria in healthy, optimally performing chicken layers across all stages of the commercial farm sequence. Consequently, the study has set the groundwork for the development of interventions that seek to enhance production performance and to prevent and control infectious diseases through the modulation of gut and respiratory bacteria.


Subject(s)
Bacteria/isolation & purification , Chickens/microbiology , Lower Gastrointestinal Tract/microbiology , Microbiota , Respiratory System/microbiology , Age Factors , Animal Husbandry , Animals , Bacteria/classification , DNA Barcoding, Taxonomic/veterinary , Gastrointestinal Microbiome , RNA, Bacterial/analysis , RNA, Ribosomal, 16S/analysis
7.
Avian Dis ; 59(2): 207-12, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26473670

ABSTRACT

Several different enteric viruses have been identified as the causes of gastrointestinal infections in poultry. Enteric virus infections are well characterized in poults, but limited studies have been conducted in older birds. The susceptibility of 2-, 7-, 12-, 30-, and 52-wk-old turkeys to turkey coronavirus (TCoV) and turkey astrovirus (TAstV) was evaluated, as well as the effect of combined infection of TAstV and TCoV in 2-wk-old poults and turkey hens. From cloacal swabs and intestines, TCoV was consistently detected by reverse transcriptase-PCR throughout the experimental period (1-21 days postinoculation [DPI]) from all age groups. In contrast, the last detection point of TAstV gradually decreased to 21, 16, and 12 DPI in birds inoculated at 2, 7, and 12 wk of age, respectively, and viral RNA was rarely detected from cloacal swabs or intestinal contents in turkey hens within 3 DPI. Infection with TAstV alone did not affect body weight in poults or egg production in hens. The combined infection of TAstV and TCoV did not induce more severe clinical signs and pathology than the TCoV infection alone. However, a severe prolonged decrease in egg production (about 50%) was observed in turkey hens in the combined infection group compared with a transient egg production drop in the TCoV-infected hens alone. The underlying mechanism regarding the age-related TAstV susceptibility and the pathogenesis of the TAstV and TCoV coinfection in layer hens needs to be further elucidated.


Subject(s)
Aging , Astroviridae Infections/veterinary , Avastrovirus/pathogenicity , Coronavirus, Turkey/pathogenicity , Enteritis, Transmissible, of Turkeys/virology , Turkeys , Animals , Antibodies, Viral/blood , Astroviridae Infections/virology , Avastrovirus/immunology , Coronavirus, Turkey/immunology , Diarrhea/veterinary , Diarrhea/virology , Female
8.
Vet World ; 17(2): 303-312, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38595666

ABSTRACT

Background and Aim: Respiratory viral infections significantly negatively impact animal welfare and have significant financial implications in the poultry industry. This study aimed to determine the frequency of the most economically relevant respiratory viruses that circulated in Egyptian chicken flocks in 2022. Materials and Methods: Chickens from 359 broiler flocks in five different Egyptian governorates in the Nile Delta (Beheira, Gharbia, Giza, Monufiya, and Qalyoubia) at marketing time (33-38 days of age) were used in this study. Combined oropharyngeal and cloacal swabs and tissue samples were collected from clinically diseased or freshly dead birds suffering from respiratory disease. Avian influenza (AI)-H5, AI-H9, Newcastle disease (ND), and infectious bronchitis virus (IBV) were analyzed by reverse transcriptase polymerase chain reaction. Results: Of the 359 flocks examined, 293 tested positive, whereas 66 were completely negative for the four viruses evaluated, with the highest positive results in Beheira. Out of 293 positive flocks, 211 were positive for a single virus, with Beheira having the highest rate, followed by Qalyoubia, Giza, and Monufiya. ND virus (NDV) was found to be the highest across all governorates, followed by IBV, AI-H9, and AI-H5. A double infection was detected in 73 flocks with either H9 or ND, or both H9 and IB could coinfect each other. The most common viral coinfections were H9 + IB, ND + IB, and ND + H9. Giza had the highest prevalence of ND + H9, H9 + IB, and ND + IB coinfection in the governorates, followed by Monufiya and Beheira. Only six out of 359 flocks were tribally infected with ND + H9 + IB in Giza, Monufiya, and Beheira governorates. On the basis of the number of flocks and the month of the year, July had the lowest number of flocks (23), while September and October had the highest number (48 flocks). Positive flock numbers were highest in October and lowest in January. Conclusion: From January to October 2022, prevalent respiratory viral infections (H5N1, NDV, H9N2, and IBV) were detected in broiler chickens across the Delta area governorate, according to the findings of the present study. In addition, IBV and H9, either alone or in combination, significantly contributed to the respiratory infection observed in broiler chickens. Regardless of the type and origin of the vaccine used, it is not possible to protect broiler chickens from the development of the infection and the subsequent dissemination of the virus into the poultry environment. In the presence of face-infectious field virus mutations, poultry vaccinations must be regularly reviewed and updated, and poultry farms must take further biosecurity measures.

9.
Acta Vet Scand ; 66(1): 27, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956671

ABSTRACT

BACKGROUND: Heterakis gallinarum (H. gallinarum) is a common poultry parasite that can be found in the ceca of many gallinaceous bird species, causing minor pathology and reduced weight gain. Most infections go unnoticed in commercial flocks due to the dependence on fecal egg counts, which are prone to false-negative diagnoses. Furthermore, there is a lack of research on gastrointestinal nematodes that use molecular identification methods, which could be essential for rapid diagnosis and developing efficient control approaches. As a result, the study aimed to look at the cause of mortality in layer chickens induced by H. gallinarum in Egyptian poultry farms using morphological, ultrastructural, and molecular characterization. Histopathological, immunohistochemical, and cell-mediated immune responses from damaged cecal tissues were also examined. RESULTS: Seventy bird samples from ten-layer flocks of different breeds (Native, white, and brown layers) suffering from diarrhea, decreased egg output, and emaciation were collected. Cecal samples were collected from affected and non-affected birds and were examined for parasitic diseases using light and a scanning electron microscope. The mitochondrial cytochrome oxidase 1 (COX1) gene was used to characterize H. gallinarum. Our results showed that the collected nematodal worms were identified as H. gallinarum (male and female), further confirmed by COX1 gene amplification and sequence alignment. Gene expression analysis of the inflammatory markers in infected tissues showed a significant up-regulation of IL-2, IFN-γ, TLR-4, and IL-1ß and a significant down-regulation of the anti-inflammatory IL-10. The mRNA level of the apoptotic cas-3 revealed apoptotic activity among the H. gallinarum samples compared to the control group. CONCLUSIONS: Our results implemented the use of molecular methods for the diagnosis of Heterakis, and this is the first report showing the tissue immune response following infection in layers: upregulation of IL-1ß, IFN-γ, Il-2, and TLR-4, while down-regulation of anti-inflammatory IL-10 in cecal tissue, Cas-3 apoptotic activity and Nuclear factor-κB (NF-κB)activity with immunophenotyping of T-cells in Heterakis infected tissue.


Subject(s)
Cecum , Chickens , Poultry Diseases , Typhlitis , Animals , Poultry Diseases/parasitology , Poultry Diseases/immunology , Poultry Diseases/pathology , Typhlitis/veterinary , Typhlitis/parasitology , Typhlitis/pathology , Cecum/parasitology , Cecum/pathology , Female , Immunity, Cellular , Ascaridida Infections/veterinary , Ascaridida Infections/parasitology , Ascaridoidea , Egypt
10.
Mol Biol Cell ; 35(5): ar62, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38507240

ABSTRACT

The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) accessory protein Orf6 works as an interferon antagonist, in part, by inhibiting the nuclear import activated p-STAT1, an activator of interferon-stimulated genes, and the export of the poly(A) RNA. Insight into the transport regulatory function of Orf6 has come from the observation that Orf6 binds to the nuclear pore complex (NPC) components: Rae1 and Nup98. To gain further insight into the mechanism of Orf6-mediated transport inhibition, we examined the role of Rae1 and Nup98. We show that Rae1 alone is not necessary to support p-STAT1 import or nuclear export of poly(A) RNA. Moreover, the loss of Rae1 suppresses the transport inhibitory activity of Orf6. We propose that the Rae1/Nup98 complex strategically positions Orf6 within the NPC where it alters FG-Nup interactions and their ability to support nuclear transport. In addition, we show that Rae1 is required for normal viral protein production during SARS-CoV-2 infection presumably through its role in supporting Orf6 function.


Subject(s)
Active Transport, Cell Nucleus , COVID-19 , Nuclear Pore , Nucleocytoplasmic Transport Proteins , SARS-CoV-2 , Humans , COVID-19/metabolism , Interferons/metabolism , Nuclear Pore/metabolism , Nuclear Pore Complex Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA, Messenger/metabolism , SARS-CoV-2/metabolism , Viral Proteins/metabolism , Nuclear Matrix-Associated Proteins/metabolism
11.
Avian Dis ; 57(3): 650-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24283132

ABSTRACT

Turkey coronavirus (TCoV) infection causes enteritis in turkeys of varying ages with high mortality in young birds. In older birds, field evidence indicates the possible involvement of TCoV in egg-production drops in turkey hens. However, no experimental studies have been conducted to demonstrate TCoV pathogenesis in turkey hens and its effect on reproductive performance. In the present study, we assessed the possible effect of TCoV on the reproductive performance of experimentally infected turkey hens. In two separate trials, 29- to 30-wk-old turkey hens in peak egg production were either mock-infected or inoculated orally with TCoV (Indiana strain). Cloacal swabs and intestinal and reproductive tissues were collected and standard reverse-transcription PCR was conducted to detect TCoV RNA. In the cloacal swabs, TCoV was detected consistently at 3, 5, 7, and 12 days postinoculation (DPI) with higher rates of detection after 5 DPI (> 90%). All intestinal samples were also positive for TCoV at 7 DPI, and microscopic lesions consisting of severe enteritis with villous atrophy were observed in the duodenum and jejunum of TCoV-infected hens. In one of the trials TCoV was detected from the oviduct of two birds at 7 DPI; however, no or mild microscopic lesions were present. In both experimental trials an average of 28%-29% drop in egg production was observed in TCoV-infected turkey hens between 4 and 7 DPI. In a separate trial we also confirmed that TCoV can efficiently transmit from infected to contact control hens. Our results show that TCoV infection can affect the reproductive performance in turkey hens, causing a transient drop in egg production. This drop in egg production most likely occurred as consequence of the severe enteritis produced by the TCoV. However, the potential replication of TCoV in the oviduct and its effect on pathogenesis should be considered and further investigated.


Subject(s)
Coronavirus, Turkey/physiology , Enteritis, Transmissible, of Turkeys/physiopathology , Reproduction , Turkeys , Animals , Cloaca/virology , Enteritis, Transmissible, of Turkeys/pathology , Enteritis, Transmissible, of Turkeys/transmission , Enteritis, Transmissible, of Turkeys/virology , Female , Intestines/virology , Ovum/virology , Reverse Transcriptase Polymerase Chain Reaction/veterinary
12.
J Adv Vet Anim Res ; 10(4): 763-772, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38370888

ABSTRACT

Objective: Aspergillosis is a disease that affects several species of birds and causes substantial losses in the poultry business. The purpose of the investigation was to identify the pathogen responsible for a respiratory outbreak among juvenile ducklings. Materials and Methods: An epidemic of Aspergillosis infected a total of 800 Muscovy ducks that were being reared in El-Beheira Governorate. Tissue samples were obtained to isolate suspected fungi from diseased birds and the hatchery environment. In addition, identification and molecular characterization were performed on the obtained fungal isolates. Results: Affected birds displayed acute respiratory manifestations such as difficulty breathing, gasping for air, nasal discharge, and a mortality rate of up to 28.1%. Postmortem examination revealed bronchitis, tracheitis, congested lungs, air sacculitis, severe multifocal granulomatous pneumonia, a congested, enlarged liver, and a congested kidney with nephritis. Mycological examination revealed seven Aspergillus (A.) spp. isolates from ducklings and six from hatcheries. Isolate colonial morphology and microscopical examination were as follows: A. fumigatus, A. niger, Syncephalastrum racemosum, and four untypable isolates. These isolates were further identified by polymerase chain reaction (PCR), and the internal transcribed spacers (ITSs) gene was detected. Four representative isolates were submitted for sequencing and further phylogenetic analysis. The source of duckling infection might be linked to the hatchery environment due to the observed similarity of isolates from both affected birds and the hatchery, as evidenced by phylogenetic analysis. Conclusion: Our findings demonstrated the significance of appropriate hatchery control in preventing infection in young ducklings. Furthermore, the use of molecular identification techniques would be helpful for tracing the source of infection and rapid diagnosis of Aspergillus in the field.

13.
J Mol Biol ; 435(16): 168170, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37271493

ABSTRACT

Argonaute 2 (Ago2) is a key component of the RNA interference (RNAi) pathway, a gene-regulatory system that is present in most eukaryotes. Ago2 uses microRNAs (miRNAs) and small interfering RNAs (siRNAs) for targeting to homologous mRNAs which are then degraded or translationally suppressed. In plants and invertebrates, the RNAi pathway has well-described roles in antiviral defense, but its function in limiting viral infections in mammalian cells is less well understood. Here, we examined the role of Ago2 in replication of the betacoronavirus SARS-CoV-2, the etiologic agent of COVID-19. Microscopic analyses of infected cells revealed that a pool of Ago2 closely associates with viral replication sites and gene ablation studies showed that loss of Ago2 resulted in over 1,000-fold increase in peak viral titers. Replication of the alphacoronavirus 229E was also significantly increased in cells lacking Ago2. The antiviral activity of Ago2 was dependent on both its ability to bind small RNAs and its endonuclease function. Interestingly, in cells lacking Dicer, an upstream component of the RNAi pathway, viral replication was the same as in parental cells. This suggests that the antiviral activity of Ago2 is independent of Dicer processed miRNAs. Deep sequencing of infected cells by other groups identified several SARS-CoV-2-derived small RNAs that bind to Ago2. A mutant virus lacking the most abundant ORF7A-derived viral miRNA was found to be significantly less sensitive to Ago2-mediated restriction. This combined with our findings that endonuclease and small RNA-binding functions of Ago2 are required for its antiviral function, suggests that Ago2-small viral RNA complexes target nascent viral RNA produced at replication sites for cleavage. Further studies are required to elucidate the processing mechanism of the viral small RNAs that are used by Ago2 to limit coronavirus replication.


Subject(s)
Argonaute Proteins , COVID-19 , MicroRNAs , RNA Interference , SARS-CoV-2 , Animals , Humans , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , COVID-19/metabolism , COVID-19/virology , MicroRNAs/genetics , RNA, Double-Stranded , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Viral/genetics , SARS-CoV-2/genetics , SARS-CoV-2/metabolism
14.
ACS Cent Sci ; 9(7): 1374-1387, 2023 Jul 26.
Article in English | MEDLINE | ID: mdl-37521792

ABSTRACT

Native mass spectrometry (nMS) screening of natural glycan libraries against glycan-binding proteins (GBPs) is a powerful tool for ligand discovery. However, as the glycan concentrations are unknown, affinities cannot be measured directly from natural libraries. Here, we introduce Concentration-Independent (COIN)-nMS, which enables quantitative screening of natural glycan libraries by exploiting slow mixing of solutions inside a nanoflow electrospray ionization emitter. The affinities (Kd) of detected GBP-glycan interactions are determined, simultaneously, from nMS analysis of their time-dependent relative abundance changes. We establish the reliability of COIN-nMS using interactions between purified glycans and GBPs with known Kd values. We also demonstrate the implementation of COIN-nMS using the catch-and-release (CaR)-nMS assay for glycosylated GBPs. The COIN-CaR-nMS results obtained for plant, fungal, viral, and human lectins with natural libraries containing hundreds of N-glycans and glycopeptides highlight the assay's versatility for discovering new ligands, precisely measuring their affinities, and uncovering "fine" specificities. Notably, the COIN-CaR-nMS results clarify the sialoglycan binding properties of the SARS-CoV-2 receptor binding domain and establish the recognition of monosialylated hybrid and biantennary N-glycans. Moreover, pharmacological depletion of host complex N-glycans reduces both pseudotyped virions and SARS-CoV-2 cell entry, suggesting that complex N-glycans may serve as attachment factors.

15.
Cells ; 10(12)2021 12 12.
Article in English | MEDLINE | ID: mdl-34944018

ABSTRACT

Mayaro virus (MAYV) is an emerging mosquito-transmitted virus that belongs to the genus Alphavirus within the family Togaviridae. Humans infected with MAYV often develop chronic and debilitating arthralgia and myalgia. The virus is primarily maintained via a sylvatic cycle, but it has the potential to adapt to urban settings, which could lead to large outbreaks. The interferon (IFN) system is a critical antiviral response that limits replication and pathogenesis of many different RNA viruses, including alphaviruses. Here, we investigated how MAYV infection affects the induction phase of the IFN response. Production of type I and III IFNs was efficiently suppressed during MAYV infection, and mapping revealed that expression of the viral non-structural protein 2 (nsP2) was sufficient for this process. Interactome analysis showed that nsP2 interacts with DNA-directed RNA polymerase II subunit A (Rpb1) and transcription initiation factor IIE subunit 2 (TFIIE2), which are host proteins required for RNA polymerase II-mediated transcription. Levels of these host proteins were reduced by nsP2 expression and during infection by MAYV and related alphaviruses, suggesting that nsP2-mediated inhibition of host cell transcription is an important aspect of how some alphaviruses block IFN induction. The findings from this study may prove useful in design of vaccines and antivirals, which are currently not available for protection against MAYV and infection by other alphaviruses.


Subject(s)
Alphavirus/metabolism , Host-Pathogen Interactions , Interferons/metabolism , Protein Subunits/metabolism , Transcription Factors, TFII/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Down-Regulation , Humans , Interferon Regulatory Factor-3/metabolism , Protein Binding , Protein Transport , RNA Polymerase II/metabolism , Transcription, Genetic
16.
Avian Dis ; 64(3): 277-285, 2020 09 01.
Article in English | MEDLINE | ID: mdl-33205170

ABSTRACT

Although poultry microbiome discoveries are increasing due to the potential impact on poultry performance, studies examining the poultry respiratory microbiome are challenging because of the low microbial biomass and uniqueness of the avian respiratory tract, making it difficult to sample enough material for microbial analysis. Invasive sampling techniques requiring euthanasia are currently used to increase microbial mass for the analysis, thus making it impossible to sample individual birds longitudinally. In this study, we compared invasive (nasal wash, upper tracheal wash, lower tracheal wash, and lower respiratory lavage) and noninvasive (tracheal and choanal swabs) respiratory sampling techniques in two independent experiments by using 4-wk-old chickens. We first established the experimental baseline of respiratory microbiota by using invasive techniques to enable reasonable comparisons between sampling methods and between experiments. Although noninvasive sampling (live-bird swabs) resulted in lower 16S ribosomal RNA gene copy numbers compared with invasive sampling, live swabs were able to detect the dominant microbes captured by invasive techniques. Nevertheless, swabs from euthanatized birds were more reflective of the microbiota captured through invasive methods than live swab. Furthermore, from two separate experiments, we also demonstrated that respiratory microbiota sampling is highly reproducible, especially in the trachea and lower respiratory tract. Our study provides new insights and perspectives on decision making when sampling and studying poultry respiratory microbiota.


Subject(s)
Bacteria/isolation & purification , Chickens/microbiology , Microbiota , Respiratory System/microbiology , Specimen Handling/veterinary , Animals , Bacteria/genetics , RNA, Bacterial/analysis , RNA, Ribosomal, 16S/analysis , Sequence Analysis, DNA/veterinary , Specimen Handling/instrumentation , Specimen Handling/methods
17.
Poult Sci ; 99(12): 6513-6524, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33248566

ABSTRACT

Toll-like receptor 3 (TLR3) induces host innate immune response on recognition of viral double-stranded RNA (dsRNA). Although several studies of avian TLR3 have been reported, none of these studies used a gene knockout (KO) model to directly assess its role in inducing the immune response and effect on other dsRNA receptors. In this study, we determined the coding sequence of quail TLR3, identified isoforms, and generated TLR3 KO quail fibroblast (QT-35) cells using a CRISPR/Cas9 system optimized for avian species. The TLR3-mediated immune response was studied by stimulating the wild-type (WT) and KO QT-35 cells with synthetic dsRNA or polyinosinic:polycytidylic acid [poly(I:C)] or infecting the cells with different RNA viruses such as influenza A virus, avian reovirus, and vesicular stomatitis virus. The direct poly(I:C) treatment significantly increased IFN-ß and IL-8 gene expression along with the cytoplasmic dsRNA receptor, melanoma differentiation-associated gene 5 (MDA5), in WT cells, whereas no changes in all corresponding genes were observed in KO cells. We further confirmed the antiviral effects of poly(I:C)-induced TLR3-mediated immunity by demonstrating significant reduction of virus titer in poly(I:C)-treated WT cells, but not in TLR3 KO cells. On virus infection, varying levels of IFN-ß, IL-8, TLR3, and MDA5 gene upregulation were observed depending on the viruses. No major differences in gene expression level were observed between WT and TLR3 KO cells, which suggests a relatively minor role of TLR3 in sensing and exerting immune response against the viruses tested in vitro. Our data show that quail TLR3 is an important endosomal dsRNA receptor responsible for regulation of type I interferon and proinflammatory cytokine, and affect the expression of MDA5, another dsRNA receptor, most likely through cytokine-mediated communication.


Subject(s)
Birds , Immunity , Protein Isoforms , Toll-Like Receptor 3 , Animals , Birds/immunology , Cells, Cultured , Fibroblasts/immunology , Immunity/immunology , Poly I-C/pharmacology , Protein Isoforms/immunology , Quail/immunology , Toll-Like Receptor 3/chemistry , Toll-Like Receptor 3/immunology
18.
Vet Microbiol ; 235: 170-179, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31383299

ABSTRACT

Turkey arthritis reovirus (TARV) infections have been recognized since 2011 to cause disease and significant economic losses to the U.S. turkey industry. Reoviral arthritis has been reproduced in commercial-origin turkeys. However, determination of pathogenesis or vaccine efficacy in these turkeys can be complicated by enteric reovirus strains and other pathogens that ubiquitously exist at subclinical levels among commercial turkey flocks. In this study, turkeys from a specific-pathogen-free (SPF) flock were evaluated for use as a turkey reoviral arthritis model. One-day-old or 1-week-old poults were orally inoculated with TARV (O'Neil strain) and monitored for disease onset and progression. A gut isolate of turkey reovirus (MN1 strain) was also tested for comparison. Disease was observed only in TARV-infected birds. Features of reoviral arthritis in SPF turkeys included swelling of hock joints, tenosynovitis, distal tibiotarsal cartilage erosion, and gait defects (lameness). Moreover, TARV infection resulted in a significant depression of body weights during the early times post-infection. Age-dependent susceptibility to TARV infection was unclear. TARV was transmitted to all sentinel birds, which manifested high levels of tenosynovitis and tibiotarsal cartilage erosion. Simulation of stressful conditions by dexamethasone treatment did not affect the viral load or exacerbate the disease. Collectively, the clinical and pathological features of reoviral arthritis in the SPF turkey model generally resembled those induced in commercial turkeys under field and/or experimental conditions. The SPF turkey reoviral arthritis model will be instrumental in evaluation of TARV pathogenesis and reoviral vaccine efficacy.


Subject(s)
Arthritis/veterinary , Disease Models, Animal , Reoviridae Infections/veterinary , Specific Pathogen-Free Organisms , Turkeys , Animals , Arthritis/virology
19.
Vaccine ; 37(43): 6454-6462, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31506195

ABSTRACT

Development of a broadly reactive influenza vaccine that can provide protection against emerging type A influenza viruses is a big challenge. We previously demonstrated that a vaccine displaying the extracellular domain of the matrix protein 2 (M2e) on the surface loops of norovirus P-particle (M2eP) can partially protect chickens against several subtypes of avian influenza viruses. In the current study, a chimeric vaccine containing a conserved peptide from the subunit 2 of hemagglutinin (HA) glycoprotein (HA2) and Arabidopsis thaliana cyanase protein (AtCYN) (HA2-AtCYN vaccine) was evaluated in 2-weeks-old chickens. Depending on the route of administration, the HA2-AtCYN vaccine was shown to induce various levels of HA2-specific IgA in tears as well as serum IgG, which were associated with partial protection of chickens against tracheal shedding of a low pathogenicity H5N2 challenge virus. Furthermore, intranasal administration with a combination of HA2-AtCYN and M2eP vaccines resulted in enhanced protection compared to each vaccine alone. Simultaneous intranasal administration of the vaccines did not interfere with secretory IgA induction by each vaccine. Additionally, significantly higher M2eP-specific proliferative responses were observed in peripheral blood mononuclear cells of all M2eP-vaccinated groups when compared with the mock-vaccinated group. Although tripling the number of M2e copies did not enhance the protective efficacy of the chimeric vaccine, it significantly reduced immunodominance of P-particle epitopes without affecting the robustness of M2e-specific immune responses. Taken together, our data suggests that mucosal immunization of chickens with combinations of mechanistically different cross-subtype-conserved vaccines has the potential to enhance the protective efficacy against influenza virus challenge.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Norovirus , Viral Matrix Proteins/immunology , Animals , Antibodies, Viral/analysis , Antibodies, Viral/blood , Arabidopsis/enzymology , Carbon-Nitrogen Lyases/genetics , Carbon-Nitrogen Lyases/immunology , Chickens/immunology , Cross Protection , Epitopes/immunology , Immunoglobulin A/analysis , Immunoglobulin G/blood , Influenza A Virus, H5N2 Subtype , Influenza Vaccines/administration & dosage , Specific Pathogen-Free Organisms , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Matrix Proteins/genetics
20.
Vaccine ; 37(10): 1356-1364, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30691981

ABSTRACT

Avian influenza in poultry continues to be a great concern worldwide, and the currently licensed inactivated influenza vaccines are not effective against the novel strains of influenza virus that continue to emerge in the field. This warrants the development of more broadly protective influenza vaccines or vaccination regimens. Live attenuated influenza vaccines (LAIVs) and subunit vaccines derived from viral peptides, such as the highly conserved ectodomain of influenza virus matrix protein 2 (M2e), can offer a more broadly reactive immune response. In chickens, we previously showed that a chimeric norovirus P particle containing M2e (M2eP) could provide partial but broad immunity, when administered as a standalone vaccine, and also enhanced the protective efficacy of inactivated vaccine when used in a combination regimen. We also demonstrated that a naturally-selected NS1-truncated H7N3 LAIV (pc4-LAIV) was highly efficacious against antigenically distant heterologous H7N2 low pathogenicity avian influenza virus challenge, especially when used as the priming vaccine in a prime-boost vaccination regimen. In this study, we investigated the cross-subtype protective efficacy of pc4-LAIV in conjunction with M2eP using single vaccination, combined treatment, and prime-boost approaches. Chickens vaccinated with pc4-LAIV showed significant reduction of tracheal shedding of a low pathogenicity H5N2 challenge virus. This cross-subtype protective efficacy was further enhanced, during the initial stages of challenge virus replication, in chickens that received a vaccination regimen consisting of priming with pc4-LAIV at 1 day of age and boosting with M2eP. Further, H5N2-specific serum IgG and pc4-LAIV-specific hemagglutination-inhibition antibody titers were enhanced in LAIV-primed and M2eP boost-vaccinated chickens. Taken together, our data point to the need of further investigation into the benefits of combined and prime-boost vaccination schemes utilizing LAIV and epitope-based vaccines, to develop more broadly protective vaccination regimens.


Subject(s)
Antibodies, Viral/blood , Cross Protection , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Norovirus , Viral Matrix Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Chickens , Hemagglutination Inhibition Tests , Immunization Schedule , Immunization, Secondary , Influenza A Virus, H5N2 Subtype , Influenza A Virus, H7N2 Subtype , Influenza A Virus, H7N3 Subtype , Influenza Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Matrix Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL