Enteric Viruses in Animals

A special issue of Viruses (ISSN 1999-4915). This special issue belongs to the section "Animal Viruses".

Deadline for manuscript submissions: closed (15 December 2020) | Viewed by 48452

Special Issue Editor


E-Mail Website
Guest Editor
National Institute of Animal Health, NARO, Ibaraki 305-0856, Japan
Interests: reverse genetics technique; enteric virus; coronavirus; rotavirus; calicivirus; VLPs (virus-like particles); anti-IgY; animal coronaviruses; animal rotaviruses; pathogenicity
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear colleagues,

The main topic of this Special Issue is enteric viruses that cause acute and severe diarrhea in animals in the agricultural and veterinary industry. Enteric viruses include mainly emerging and re-emerging Coronaviridae (PEDV, TGEV, PDCoV, SADS-CoV, Bovine CoV, Feline CoV, and Torovirus), Reoviridae (RVA, RVB, RVC and RVH, and Mammalian orthoreovirus), and Caliciviridae (Norovirus and Sapovirus). Diarrhea due to enteric viruses leads to deterioration of health, insufficient body weight gain, and deaths of young animals, resulting in huge economic losses. However, available information is still limited regarding enteric viruses, and hence, there are few effective strategies for control and prevention of enteric viruses, despite their significant economic impact. Recently, it has become possible to utilize reverse genetic approaches and cultivation in culture (enteroid and iPS) for these enteric viruses, which will prompt further elucidation of their molecular mechanisms and facilitate the development of treatment. Therefore, this Special Issue welcomes all types of manuscripts (e.g., reviews, research articles, and short communications), including novel findings with respect to molecular mechanisms, pathogenicity, host–virus interaction, cultivation in culture, and treatment of enteric viruses.

Dr. Tohru Suzuki
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Viruses is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2600 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • coronaviruses
  • rotaviruses
  • caliciviruses
  • reverse genetics
  • cultivation
  • pathogenicity
  • treatment

Published Papers (10 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

15 pages, 6705 KiB  
Article
Chimeric Porcine Deltacoronaviruses with Sparrow Coronavirus Spike Protein or the Receptor-Binding Domain Infect Pigs but Lose Virulence and Intestinal Tropism
by Xiaoyu Niu, Yixuan J. Hou, Kwonil Jung, Fanzhi Kong, Linda J. Saif and Qiuhong Wang
Viruses 2021, 13(1), 122; https://doi.org/10.3390/v13010122 - 17 Jan 2021
Cited by 11 | Viewed by 3344
Abstract
Porcine deltacoronavirus (PDCoV) strain OH-FD22 infects poultry and shares high nucleotide identity with sparrow-origin deltacoronaviruses (SpDCoV) ISU73347 and HKU17 strains. We hypothesized that the spike (S) protein or receptor-binding domain (RBD) from these SpDCoVs would alter the host and tissue tropism of PDCoV. [...] Read more.
Porcine deltacoronavirus (PDCoV) strain OH-FD22 infects poultry and shares high nucleotide identity with sparrow-origin deltacoronaviruses (SpDCoV) ISU73347 and HKU17 strains. We hypothesized that the spike (S) protein or receptor-binding domain (RBD) from these SpDCoVs would alter the host and tissue tropism of PDCoV. First, an infectious cDNA clone of PDCoV OH-FD22 strain (icPDCoV) was generated and used to construct chimeric icPDCoVs harboring the S protein of HKU17 (icPDCoV-SHKU17) or the RBD of ISU73347 (icPDCoV-RBDISU). To evaluate their pathogenesis, neonatal gnotobiotic pigs were inoculated orally/oronasally with the recombinant viruses or PDCoV OH-FD22. All pigs inoculated with icPDCoV or OH-FD22 developed severe diarrhea and shed viral RNA at moderate-high levels (7.62–10.56 log10 copies/mL) in feces, and low-moderate levels in nasal swabs (4.92–8.48 log10 copies/mL). No pigs in the icPDCoV-SHKU17 and icPDCoV-RBDISU groups showed clinical signs. Interestingly, low-moderate levels (5.07–7.06 log10 copies/mL) of nasal but not fecal viral RNA shedding were detected transiently at 1–4 days post-inoculation in 40% (2/5) of icPDCoV-SHKU17- and 50% (1/2) of icPDCoV-RBDISU-inoculated pigs. These results confirm that PDCoV infected both the upper respiratory and intestinal tracts of pigs. The chimeric viruses displayed an attenuated phenotype with the loss of tropism for the pig intestine. The SpDCoV S protein and RBD reduced viral replication in pigs, suggesting limited potential for cross-species spillover upon initial passage. Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
Show Figures

Figure 1

12 pages, 281 KiB  
Article
Comparison of Four Commercially Available Point-of-Care Tests to Detect Antibodies against Canine Parvovirus in Dogs
by Michèle Bergmann, Mike Holzheu, Yury Zablotski, Stephanie Speck, Uwe Truyen, Reinhard K. Straubinger and Katrin Hartmann
Viruses 2021, 13(1), 18; https://doi.org/10.3390/v13010018 - 23 Dec 2020
Cited by 7 | Viewed by 2555
Abstract
Measuring antibodies to evaluate dogs’ immunity against canine parvovirus (CPV) is useful to avoid unnecessary re-vaccinations. The study aimed to evaluate the quality and practicability of four point-of-care (POC) tests for detection of anti-CPV antibodies. The sera of 198 client-owned and 43 specific [...] Read more.
Measuring antibodies to evaluate dogs’ immunity against canine parvovirus (CPV) is useful to avoid unnecessary re-vaccinations. The study aimed to evaluate the quality and practicability of four point-of-care (POC) tests for detection of anti-CPV antibodies. The sera of 198 client-owned and 43 specific pathogen-free (SPF) dogs were included; virus neutralization was the reference method. Specificity, sensitivity, positive and negative predictive value (PPV and NPV), and overall accuracy (OA) were calculated. Specificity was considered to be the most important indicator for POC test performance. Differences between specificity and sensitivity of POC tests in the sera of all dogs were determined by McNemar, agreement by Cohen’s kappa. Prevalence of anti-CPV antibodies in all dogs was 80% (192/241); in the subgroup of client-owned dogs, it was 97% (192/198); and in the subgroup of SPF dogs, it was 0% (0/43). FASTest® and CanTiCheck® were easiest to perform. Specificity was highest in the CanTiCheck® (overall dogs, 98%; client-owned dogs, 83%; SPF dogs, 100%) and the TiterCHEK® (overall dogs, 96%; client-owned dogs, 67%; SPF dogs, 100%); no significant differences in specificity were observed between the ImmunoComb®, the TiterCHEK®, and the CanTiCheck®. Sensitivity was highest in the FASTest® (overall dogs, 95%; client-owned dogs, 95%) and the CanTiCheck® (overall dogs, 80%; client-owned dogs, 80%); sensitivity of the FASTest® was significantly higher compared to the one of the other three tests (McNemars p-value in each comparison: <0.001). CanTiCheck® would be the POC test of choice when considering specificity and practicability. However, differences in the number of false positive results between CanTiCheck®, TiterCHEK®, and ImmunoComb® were minimal. Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
18 pages, 4008 KiB  
Article
Development of Genotype-Specific Anti-Bovine Rotavirus A Immunoglobulin Yolk Based on a Current Molecular Epidemiological Analysis of Bovine Rotaviruses A Collected in Japan during 2017–2020
by Koki Odagiri, Nobuki Yoshizawa, Hisae Sakihara, Koji Umeda, Shofiqur Rahman, Sa Van Nguyen and Tohru Suzuki
Viruses 2020, 12(12), 1386; https://doi.org/10.3390/v12121386 - 03 Dec 2020
Cited by 6 | Viewed by 2710
Abstract
Bovine rotavirus A (RVA), a major causative pathogen of diarrhea in dairy and Japanese beef calves, has led to severe economic losses in numerous countries. A dual genotyping system based on genomic segments encoding VP7 (G genotype) and VP4 (P genotype), comprising the [...] Read more.
Bovine rotavirus A (RVA), a major causative pathogen of diarrhea in dairy and Japanese beef calves, has led to severe economic losses in numerous countries. A dual genotyping system based on genomic segments encoding VP7 (G genotype) and VP4 (P genotype), comprising the outer layer of the virion, has been used to understand the epidemiological dynamics of RVAs at the national and global levels. This study aimed to investigate occurrence frequency of G and P genotypes for multiple bovine RVAs from calf diarrheic samples collected in Japan from 2017 to 2020. After we produced anti-bovine RVA immunoglobulin yolks (IgYs) from hens immunized with the two RVAs with different genotypes (G6P[5] and G10P[11]) selected on the basis of the current epidemiological survey, we investigated cross-reactivity against bovine RVAs with different G and P combinations owing to establish a useful strategy to protect calves from RVA infections using the two IgYs. Consequently, the two produced anti-bovine IgYs showed strong cross-reactivity against bovine RVAs with the same G and/or P genotypes in neutralization assay, respectively. Therefore, our data suggest the possibility of a passive immunization to protect calves from a bovine RVA infections epidemic in Japan via oral administration of the two IgYs into calves. The findings presented herein will provide important information that IgY is one of the effective tools to prevent infections of various pathogens. Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
Show Figures

Figure 1

14 pages, 2490 KiB  
Article
Deletion in the S1 Region of Porcine Epidemic Diarrhea Virus Reduces the Virulence and Influences the Virus-Neutralizing Activity of the Antibody Induced
by Kuo-Jung Tsai, Ming-Chung Deng, Fun-In Wang, Shu-Hui Tsai, Chieh Chang, Chia-Yi Chang and Yu-Liang Huang
Viruses 2020, 12(12), 1378; https://doi.org/10.3390/v12121378 - 02 Dec 2020
Cited by 8 | Viewed by 2462
Abstract
Porcine epidemic diarrhea virus (PEDV) causes severe diarrhea and a high rate of mortality in suckling pigs. The epidemic of PEDV that occurred after 2013 was caused by non-insertion and deletion of S gene (S-INDEL) PEDV strains. During this epidemic, a variant of [...] Read more.
Porcine epidemic diarrhea virus (PEDV) causes severe diarrhea and a high rate of mortality in suckling pigs. The epidemic of PEDV that occurred after 2013 was caused by non-insertion and deletion of S gene (S-INDEL) PEDV strains. During this epidemic, a variant of the non-S-INDEL PEDV strain with a large deletion of 205 amino acids on the spike gene (5-17-V) was also found to co-exist with a non-S-INDEL PEDV without deletion (5-17-O). Herein, we describe the differences in the complete genome, distribution, virulence, and antigenicity between strain 5-17-O and variant strain 5-17-V. The deletion of 205 amino acids was primarily located in the S1O domain and was associated with milder clinical signs and lower mortality in suckling pigs than those of the 5-17-O strain. The 5-17-V strain-induced antibody did not completely cross-neutralize the 5-17-O strain. In conclusion, the deletion in the S1 region reduces the virulence of PEDV and influences the virus-neutralizing activities of the antibody it induces. Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
Show Figures

Figure 1

10 pages, 1119 KiB  
Article
Application of a Photocatalyst as an Inactivator of Bovine Coronavirus
by Nobuki Yoshizawa, Ryoko Ishihara, Daisuke Omiya, Midori Ishitsuka, Shouichirou Hirano and Tohru Suzuki
Viruses 2020, 12(12), 1372; https://doi.org/10.3390/v12121372 - 30 Nov 2020
Cited by 23 | Viewed by 2941
Abstract
Bovine coronavirus (BCoV), a major causative pathogen of bovine enteric and respiratory diseases and a zoonotic pathogen transmissible between animals and humans, has led to severe economic losses in numerous countries. BCoV belongs to the genus Betacoronavirus, which is a model of [...] Read more.
Bovine coronavirus (BCoV), a major causative pathogen of bovine enteric and respiratory diseases and a zoonotic pathogen transmissible between animals and humans, has led to severe economic losses in numerous countries. BCoV belongs to the genus Betacoronavirus, which is a model of a pathogen that is threatening human health and includes severe acute respiratory syndrome coronavirus (SARS-CoV), SARS-CoV-2, and Middle East respiratory syndrome coronavirus. This study aimed to determine whether photocatalytic material effectively reduces CoVs in the environment. Using the film adhesion method of photocatalytic materials, we assessed its antiviral activity and the effect of visible light irradiation according to methods defined by the International Organization for Standardization. Consequently, photocatalytic material was found to have antiviral activity, reducing the viral loads by 2.7 log TCID50 (tissue culture infective dose 50)/0.1 mL (500 lux), 2.8 log TCID50/0.1 mL (1000 lux), and 2.4 log TCID50/0.1 mL (3000 lux). Hence, this photocatalytic material might be applicable not only to reducing CoVs in the cattle breeding environment but also perhaps in other indoor spaces, such as offices and hospital rooms. To our knowledge, this study is the first to evaluate the antiviral activity of a photocatalytic material against CoV. Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
Show Figures

Figure 1

12 pages, 2319 KiB  
Article
Fatal Interstitial Pneumonia Associated with Bovine Coronavirus in Cows from Southern Italy
by Maria Grazia Amoroso, Giuseppe Lucifora, Barbara Degli Uberti, Francesco Serra, Giovanna De Luca, Giorgia Borriello, Alessandro De Domenico, Sergio Brandi, Maria Concetta Cuomo, Francesca Bove, Marita Georgia Riccardi, Giorgio Galiero and Giovanna Fusco
Viruses 2020, 12(11), 1331; https://doi.org/10.3390/v12111331 - 19 Nov 2020
Cited by 9 | Viewed by 2701
Abstract
An outbreak of winter dysentery, complicated by severe respiratory syndrome, occurred in January 2020 in a high production dairy cow herd located in a hilly area of the Calabria region. Of the 52 animals belonging to the farm, 5 (9.6%) died with severe [...] Read more.
An outbreak of winter dysentery, complicated by severe respiratory syndrome, occurred in January 2020 in a high production dairy cow herd located in a hilly area of the Calabria region. Of the 52 animals belonging to the farm, 5 (9.6%) died with severe respiratory distress, death occurring 3–4 days after the appearance of the respiratory signs (caught and gasping breath). Microbiological analysis revealed absence of pathogenic bacteria whilst Real-time PCR identified the presence of RNA from Bovine Coronavirus (BCoV) in several organs: lungs, small intestine (jejunum), mediastinal lymph nodes, liver and placenta. BCoV was therefore hypothesized to play a role in the lethal pulmonary infection. Like the other CoVs, BCoV is able to cause different syndromes. Its role in calf diarrhea and in mild respiratory disease is well known: we report instead the involvement of this virus in a severe and fatal respiratory disorder, with symptoms and disease evolution resembling those of Severe Acute Respiratory Syndromes (SARS). Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
Show Figures

Figure 1

19 pages, 5703 KiB  
Article
Porcine Epidemic Diarrhea Virus nsp15 Antagonizes Interferon Signaling by RNA Degradation of TBK1 and IRF3
by Yang Wu, Hongling Zhang, Zhaorong Shi, Jianfei Chen, Mingwei Li, Hongyan Shi, Da Shi, Longjun Guo and Li Feng
Viruses 2020, 12(6), 599; https://doi.org/10.3390/v12060599 - 31 May 2020
Cited by 33 | Viewed by 4063
Abstract
Porcine epidemic diarrhea virus (PEDV) causes a porcine disease associated with swine epidemic diarrhea. The type I interferon (IFN-I or IFN α/β) is a key mediator of innate antiviral response during virus infection. Different antagonistic strategies have been identified and determined as to [...] Read more.
Porcine epidemic diarrhea virus (PEDV) causes a porcine disease associated with swine epidemic diarrhea. The type I interferon (IFN-I or IFN α/β) is a key mediator of innate antiviral response during virus infection. Different antagonistic strategies have been identified and determined as to how PEDV infection inhibits the host’s IFN responses to escape the host innate immune pathway, but the pathogenic mechanisms of PEDV infection are not fully elucidated. Our preliminary results revealed that endogenous TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3), the key components in the IFN signaling pathway were downregulated in PEDV infected IPEC-J2 cells by iTRAQ analysis. In this study, we screened nsp15 as the most important viral encoded protein involved in TBK1 and IRF3 reduction. Endoribonuclease (EndoU) activity has been well determined for coronavirus nsp15. Three residues (H226, H241, and K282) of PEDV nsp15 were identified as critical amino acids for PEDV EndoU but not D265, which was not well correlated with published results of other coronaviruses, such as severe acute respiratory syndrome virus (SARS-CoV). Moreover, PEDV nsp15 can directly degrade the RNA levels of TBK1 and IRF3 dependent on its EndoU activity to suppress IFN production and constrain the induction of IFN stimulated genes (ISGs), by which PEDV antagonizes the host innate response to facilitate its replication. Collectively, these results have confirmed that PEDV nsp15 was capable of subverting the IFN response by the RNA degradation of TBK1 and IRF3. Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
Show Figures

Figure 1

15 pages, 2326 KiB  
Article
Virome of a Feline Outbreak of Diarrhea and Vomiting Includes Bocaviruses and a Novel Chapparvovirus
by Yanpeng Li, Emilia Gordon, Amanda Idle, Eda Altan, M. Alexis Seguin, Marko Estrada, Xutao Deng and Eric Delwart
Viruses 2020, 12(5), 506; https://doi.org/10.3390/v12050506 - 04 May 2020
Cited by 36 | Viewed by 9150
Abstract
An unexplained outbreak of feline diarrhea and vomiting, negative for common enteric viral and bacterial pathogens, was subjected to viral metagenomics and PCR. We characterized from fecal samples the genome of a novel chapparvovirus we named fechavirus that was shed by 8/17 affected [...] Read more.
An unexplained outbreak of feline diarrhea and vomiting, negative for common enteric viral and bacterial pathogens, was subjected to viral metagenomics and PCR. We characterized from fecal samples the genome of a novel chapparvovirus we named fechavirus that was shed by 8/17 affected cats and identified three different feline bocaviruses shed by 9/17 cats. Also detected were nucleic acids from attenuated vaccine viruses, members of the normal feline virome, viruses found in only one or two cases, and viruses likely derived from ingested food products. Epidemiological investigation of disease signs, time of onset, and transfers of affected cats between three facilities support a possible role for this new chapparvovirus in a highly contagious feline diarrhea and vomiting disease. Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
Show Figures

Figure 1

Review

Jump to: Research, Other

30 pages, 2768 KiB  
Review
Recent Progress in Torovirus Molecular Biology
by Makoto Ujike and Fumihiro Taguchi
Viruses 2021, 13(3), 435; https://doi.org/10.3390/v13030435 - 08 Mar 2021
Cited by 5 | Viewed by 3515
Abstract
Torovirus (ToV) has recently been classified into the new family Tobaniviridae, although it belonged to the Coronavirus (CoV) family historically. ToVs are associated with enteric diseases in animals and humans. In contrast to CoVs, which are recognised as pathogens of veterinary and medical [...] Read more.
Torovirus (ToV) has recently been classified into the new family Tobaniviridae, although it belonged to the Coronavirus (CoV) family historically. ToVs are associated with enteric diseases in animals and humans. In contrast to CoVs, which are recognised as pathogens of veterinary and medical importance, little attention has been paid to ToVs because their infections are usually asymptomatic or not severe; for a long time, only one equine ToV could be propagated in cultured cells. However, bovine ToVs, which predominantly cause diarrhoea in calves, have been detected worldwide, leading to economic losses. Porcine ToVs have also spread globally; although they have not caused serious economic losses, coinfections with other pathogens can exacerbate their symptoms. In addition, frequent inter- or intra-recombination among ToVs can increase pathogenesis or unpredicted host adaptation. These findings have highlighted the importance of ToVs as pathogens and the need for basic ToV research. Here, we review recent progress in the study of ToV molecular biology including reverse genetics, focusing on the similarities and differences between ToVs and CoVs. Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
Show Figures

Figure 1

Other

Jump to: Research, Review

10 pages, 1447 KiB  
Case Report
Rapid Resolution of Non-Effusive Feline Infectious Peritonitis Uveitis with an Oral Adenosine Nucleoside Analogue and Feline Interferon Omega
by Diane D. Addie, Johanna Covell-Ritchie, Oswald Jarrett and Mark Fosbery
Viruses 2020, 12(11), 1216; https://doi.org/10.3390/v12111216 - 27 Oct 2020
Cited by 16 | Viewed by 13933
Abstract
This is the first report of a successful treatment of a non-effusive feline infectious peritonitis (FIP) uveitis case using an oral adenosine nucleoside analogue drug and feline interferon omega, and alpha-1 acid glycoprotein (AGP) as an indicator of recovery. A 2-year-old male neutered [...] Read more.
This is the first report of a successful treatment of a non-effusive feline infectious peritonitis (FIP) uveitis case using an oral adenosine nucleoside analogue drug and feline interferon omega, and alpha-1 acid glycoprotein (AGP) as an indicator of recovery. A 2-year-old male neutered Norwegian Forest Cat presented with uveitis, keratic precipitates, mesenteric lymphadenopathy and weight loss. The cat was hypergammaglobulinaemic and had a non-regenerative anaemia. Feline coronavirus (FCoV) RNA was detected in a mesenteric lymph node fine-needle aspirate by a reverse-transcriptase polymerase chain reaction—non-effusive FIP was diagnosed. Prednisolone acetate eye drops were administered three times daily for 2 weeks. Oral adenosine nucleoside analogue (Mutian) treatment started. Within 50 days of Mutian treatment, the cat had gained over one kilogram in weight, his globulin level reduced from 77 to 51 g/L and his haematocrit increased from 22 to 35%; his uveitis resolved and his sight improved. Serum AGP level reduced from 3100 to 400 μg/mL (within normal limits). Symmetric dimethylarginine (SDMA) was above normal at 28 μg/dL, reducing to 14 μg/dL on the cessation of treatment; whether the SDMA increase was due to FIP lesions in the kidney or Mutian is unknown. Mutian treatment stopped and low-dose oral recombinant feline interferon omega begun—the cat’s recovery continued. Full article
(This article belongs to the Special Issue Enteric Viruses in Animals)
Show Figures

Figure 1

Back to TopTop