Leta i den här bloggen

fredag 13 mars 2020

Miten koronavirus toimii suolen microbiomissa? Löytyykö aiheesta artikkelia

https://www.ncbi.nlm.nih.gov/pubmed/?term=Coronavirus+in+gut+microbiome

Search results

Items: 12

1. Orthocoronavirinae, Alphacoronavirus,Tegacovirus, Feline coronaviruses (FCoV) , Canine coronaviruses

Paul A, Stayt J.
Aust Vet J. 2019 Oct;97(10):418-421. doi: 10.1111/avj.12867.
Abstract
This study reports the prevalence of potential faecal pathogens in the microbiome detected in a cohort of cats and dogs with diarrhoea in Perth, Western Australia. Records from a commercial diagnostic laboratory using faecal PCR testing between July 2014 and August 2015 were reviewed
 Of 289 feline faecal samples reviewed,
Salmonella spp. (1.7%),
Campylobacter spp. (47.6%),
Clostridium perfringens (81.3%),
Giardia spp. (11.1%),
Toxoplasma gondii (1.2%),
Tritrichomonas foetus (4.8%),
 panleukopenia virus (6.5%) and  
coronavirus (39.5%) were detected.
In dogs,
Salmonella spp. (5.4%),
Campylobacter spp. (36.3%),
 C. perfringens (85.4%),
Giardia spp. (6.2%),
parvovirus (9.4%),
 coronavirus (4.7%) and
distemper virus (1.5%) were detected.

2.Orthocoronavirinae, Alphacoronavirus, Tegacovirus, FCoV , Feline  coronavirus

Meazzi S, Stranieri A, Lauzi S, Bonsembiante F, Ferro S, Paltrinieri S, Giordano A.
Res Vet Sci. 2019 Aug;125:272-278. doi: 10.1016/j.rvsc.2019.07.003. Epub 2019 Jul 9.
Feline coronaviruses (FCoV) colonize the intestinal tract, however, due to not fully understood mutations (Feline enteric CoV strain, FeCV) , they can spread systemically and cause feline infectious peritonitis (FIP). Recent studies on human medicine report that gut microbiota is involved in the development of systemic disorders and could influence the immune response to viral diseases.
The aim of this study was to provide preliminary data on the fecal microbiota composition in healthy cats compared to FCoV-infected cats, with and without FIP. Cats were equally grouped as healthy FCoV-negative, healthy FCoV-positive or FIP affected (total n = 15). Fecal sample were evaluated for the microbiota composition. A total of 3,231,916 sequences were analyzed. The samples' alpha-diversity curves did not reach a proper plateau and, for the beta-diversity, the samples seemed not to group perfectly by category, even if the healthy FCoV-positive group showed a hybrid microbial composition between FCoV-negative and FIP groups. Although there were no taxa significantly linked to the different conditions, some peculiar patterns were recognized:
Firmicutes was always the most represented phylum, followed by
 Bacteroidetes and
Actinobacteria.
In FCoV-positive cats, the Firmicutes and Bacteroidetes were respectively over- and under-represented, compared to the other groups.
Among FIP cats, three subjects shared a similar microbiome, one cat showed a different microbial profile and the other one had the lowest number of diverse phyla. Despite the limited number of animals, some differences in the fecal microbiome between the groups were observed, suggesting to further investigate the possible correlation between gut microbiota and FCoV infection in cats.
Feline coronavirus; Feline infectious peritonitis; Gut microbiota
Free Article

3.   Orthocoronavirinae, Alpha coronavirus, Pedacovirus, PDEV, Porcine epidemic diarrhea virus

Tan Z, Dong W, Ding Y, Ding X, Zhang Q, Jiang L.
PLoS One. 2019 Jul 16;14(7):e0219868. doi: 10.1371/journal.pone.0219868. eCollection 2019.
Diarrhea, caused by porcine epidemic diarrhea virus (PEDV), is a catastrophic gastrointestinal disease among suckling piglets, with high infectivity, morbidity, and mortality, causing huge economic losses to the pig industry. In the present study, we investigated the different microbiota from the cecal mucosa and cecal contents between healthy and PEDV-infected piglets. High-throughput 16S rRNA gene sequencing was performed to explore differences. The results revealed that microbial dysbiosis by PEDV infection occurred in the cecal mucosa and contents of suckling piglets at each microbial taxonomic level. The abundance of pathogenic bacteria associated with diseases, including diarrhea, was increased. 
The abundance of Fusobacterium was 26.71% and 33.91% in cecal mucosa and contents of PEDV-infected group, respectively, whereas that in the healthy groups was 17.85% and 9.88%. 
The proportion of Proteobacteria in the infected groups was relatively high (24.67% and 22.79%, respectively), whereas that in the healthy group was 13.13% and 11.34% in the cecal mucosa and contents, respectively.
 Additionally, the proportion of Bacteroidetes in the healthy group (29.89%, 37.32%) was approximately twice that of the PEDV-infected group (15.50%, 15.39%). 
"Nitrate reduction",
"Human pathogens diarrhea", 
"Human pathogens gastroenteritis",
 "Nitrite respiration", and 
"Nitrite ammonification" were the enriched functional annotation terms in the PEDV-infected groups. Porcine epidemic diarrhea virus infection increased the proportion of harmful bacteria and    in the cecal mucosa and contents of suckling piglets. Our findings suggest that determining the intestinal microbiota might provide a promising method to prevent PEDV and open a new avenue for future research.Free PMC Article

4. Orthocoronavirinae, Gammacoronavirus, Igacovirus, Infectious Bronchitis Virus, IBV

Wu C, Yang Z, Song C, Liang C, Li H, Chen W, Lin W, Xie Q.
Poult Sci. 2018 Nov 1;97(11):3837-3846. doi: 10.3382/ps/pey268 Abstract
Yeast nucleotides are a fine functional additive in human and animals. The effects of dietary yeast nucleotides supplementation on intestinal development, expression of intestinal barrier-related genes, intestinal microbiota, and infectious bronchitis virus (IBV) antibody titer of specific pathogen-free (SPF) chickens were investigated. A total of 60 1-d-old chickens were divided into 4 groups, each of which included 3 replicates of 5 chickens. Group 1 served as a control that was fed a basal diet. Groups 2 to 4 were fed the basal diet supplemented with 0.1%, 0.3% and 0.5% yeast nucleotides, respectively. All chickens were inoculated intranasally with inactivated IBV vaccine at day 1 and day 10. At day 17, the intestinal development, expression of intestinal barrier-related genes and microbiota were evaluated. There was a significant increased ileal villus height and villus height to crypt depth ratio in group 2 (P < 0.05). Moreover, group 4 exhibited higher expression of zonula occludens-1 (ZO-1) and Occludin gene in ileum (P < 0.05), whereas groups 2 and 3 exhibited higher expression of Mucin 2 (MUC2) and trefoil factor 2 (TFF2) gene (P < 0.05), group 2 showed lower expression of IFN-α gene (P < 0.05). Dietary yeast nucleotides increased intestinal bacterial diversity (P < 0.05), and the abundance of Lactobacillus (P < 0.05). At day 10, 17, 24, 31, 38, and 45, the serum IBV antibody titers were tested. Group 2 exhibited higher IBV antibody titer at day 17 (P < 0.05), furthermore, groups 2 to 4 reached the effective levels 1 wk earlier than control group. In conclusion, dietary yeast nucleotides supplementation can help birds to mount a faster and stronger antibody response to IBV vaccine. In addition, dietary yeast nucleotides supplementation can also promote the intestinal development and barrier-related genes expression, and diversity and richness of intestinal microbiota.
Free Article

5. Nidovirales, Cornidovirinae, Coronaviridae Orthocoronavirinae, Alphacoronavirus, Tegacovirus, TGEV, Transmissible gastroenteritis virus

Xia L, Yang Y, Wang J, Jing Y, Yang Q.
Virol J. 2018 Jun 19;15(1):102. doi: 10.1186/s12985-018-1012-9.
BACKGROUND:
Pig diarrhea causes high mortality and large economic losses in the swine industry. Transmissible gastroenteritis virus (TGEV) causes pig diarrhea, with 100% mortality in piglets less than 2 weeks old. No investigation has yet been made of the small intestine of piglets that survived infection by TGEV.
METHODS:
In this study, we evaluated the impact of TGEV infection on the small intestine of recovered pigs.
RESULTS:
Histological analyses showed that TGEV infection led to villi atrophy, and reduced villous height and crypt depth. The number of SIgA positive cells, CD3+T cells, and dendritic cells (DCs) in jejunum decreased after TGEV infection in vivo. In contrast, microfold cell (M cell) numbers and cell proliferation increased in infected pigs. TGEV infection also significantly enhanced the mRNA expression levels of cytokine IL-1β, IL-6, TNF-α, IL-10, and TGF-β. Additionally, lower gene copy numbers of Lactobacillus, and higher numbers of Enterobacteriaceae, were detected in mucosal scraping samples from TGEV-infected pigs.
CONCLUSIONS:
TGEV infection damages the small intestine, impairs immune functions, and increases pathogenic bacterial loading, all of which may facilitate secondary infections by other pathogens. These findings help quantify the impact of TGEV infection and clarify the pathogenic mechanisms underlying its effects in pigs.  (Suolistomuutoket ohutsuolessa muistuttavat  keliakiamuutoksia, kommenttini. Tämä tutkimus ei paljasta, ovatko muutokset ohimeneviä vai pysyviä)
Free PMC Article

6. Nidovirales, Cornidovirinae,Coronaviridae Orthocoronavirinae,Gammacoronavirus,  Igacovirus, Duck coronaviruses, Avian coronaviruses

Vibin J, Chamings A, Collier F, Klaassen M, Nelson TM, Alexandersen S.
Sci Rep. 2018 Jun 6;8(1):8686. doi: 10.1038/s41598-018-26851-1.
We present an optimised metagenomics method for detection and characterisation of all virus types including single and double stranded DNA/RNA and enveloped and non-enveloped viruses. Initial evaluation included both spiked and non-spiked bird faecal samples as well as non-spiked human faecal samples. From the non-spiked bird samples (Australian Muscovy duck and Pacific black ducks) we detected 21 viruses, and we also present a summary of a few viruses detected in human faecal samples. We then present a detailed analysis of selected virus sequences in the avian samples that were somewhat similar to known viruses, and had good quality (Q20 or higher) and quantity of next-generation sequencing reads, and was of interest from a virological point of view, for example, avian coronavirus and avian paramyxovirus 6. Some of these viruses were closely related to known viruses while others were more distantly related with 70% or less identity to currently known/sequenced viruses. Besides detecting viruses, the technique also allowed the characterisation of host mitochondrial DNA present and thus identifying host species, while ribosomal RNA sequences provided insight into the "ribosomal activity microbiome"; of gut parasites; and of food eaten such as plants or insects, which we correlated to non-avian host associated viruses.Free PMC Article

7. Nidovirales, Cornidovirinae,Coronaviridae Orthocoronavirinae, Alphacoronavirus, Pedacovirus, PEDV, Porcine epidemic diarrhea virus

Huang MZ, Wang SY, Wang H, Cui DA, Yang YJ, Liu XW, Kong XJ, Li JY.
PLoS One. 2018 Feb 15;13(2):e0192992. doi: 10.1371/journal.pone.0192992. eCollection 2018.
Porcine epidemic diarrhea, a disastrous gastrointestinal disease, causes great financial losses due to its high infectivity, morbidity and mortality in suckling piglets despite the development and application of various vaccines. In this study, high-throughput sequencing was used to explore differences in the intestinal microbiota between uninfected piglets and piglets infected with porcine epidemic diarrhea virus (PEDV). The results revealed that the small intestinal microbiota of suckling piglets infected with PEDV showed low diversity and was dominated by Proteobacteria (49.1%). Additionally, the composition of the small intestinal microbiota of sucking piglets infected with PEDV showed marked differences from that of the uninfected piglets. Some of the taxa showing differences in abundance between uninfected piglets and piglets infected with PEDV were associated with cellular transport and catabolism, energy metabolism, the biosynthesis of other secondary metabolites, and amino acid metabolism as determined through the prediction of microbial function based on the bacterial 16S rRNA gene. Therefore, adjusting the intestinal microbiota might be a promising method for the prevention or treatment of PEDV.Free PMC Article
8. Nidovirales, Cornidovirinae,Coronaviridae, Orthocoronavirinae, Alphacoronavirus, Tegacovirus, Swine enteric CoV, Pedacovirus PEDV,  Procine epidemic diarrhea virus 

Song D, Peng Q, Chen Y, Zhou X, Zhang F, Li A, Huang D, Wu Q, Ye Y, He H, Wang L, Tang Y.
Sci Rep. 2017 Dec 12;7(1):17439. doi: 10.1038/s41598-017-17830-z.
Porcine epidemic diarrhea virus (PEDV) is a devastating cause of diarrhea in pigs worldwide. Most of studies have focused on molecular and pathogenic characterization of PEDV, whereas there were limited studies in understanding the role of gut microbiota (GM) in viral-associated diarrhea. Here, using the Illumina MiSeq platform, we examined and compared the impact of PEDV infection on the GM of sows and their piglets less than 10 days old. Our results showed that PEDV caused alternations in the structure and abundance of GM from levels of phylum to genus, and even species. For sows, a significant decrease of observed species was found in diarrheal sows than that in healthy sows (p < 0.05). The unweighted and weighted UniFrac distances also revealed considerable segregations of GM structure among healthy, asymptomatic, and diarrheal sows.
 For piglets, Bacteroidetes, the dominant bacteria in healthy piglets, were replaced by Firmicutes in asymptomatic and diarrheal piglets. The abundances of Fusobacteria and Proteobacteria were also remarkably increased in asymptomatic piglets and diarrheal piglets when compared to those of the healthy piglets. Our findings demonstrated that PEDV infection caused severe perturbations of GM, reduced probiotic bacteria, and enriched pathogenic bacteria.Free PMC Article
9. Nidovirales, Tornidovirinae, Tobaniviridae, Torovirinae, Renitovirus, Bovine Torovirus
Gomez DE, Weese JS.
Can Vet J. 2017 Dec;58(12):1267-1274. Review.
A complex community of bacteria, viruses, fungi, protists, and other microorganisms inhabit the gastrointestinal tract of calves and play important roles in gut health and disease. The viral component of the microbiome (the virome) is receiving increasing attention for its role in neonatal calf diarrhea (NCD). Rotavirus and coronavirus  (Cornidovirinae, Nidovirales)  have for a long time been associated with NCD and commercial vaccines have been produced against these agents. Recently, several other viruses which may play a role in diarrhea have been discovered in calf fecal samples, mostly by sequence-based methods. These viruses include torovirus (Tornidovirinae, Nidovirales), norovirus, nebovirus, astrovirus, kobuvirus, and enterovirus. Most studies have involved epidemiologic investigations seeking to show association with diarrhea for each virus alone or in combination with potential pathogens. However, determining the contribution of these viruses to calf diarrhea has been challenging and much uncertainty remains concerning their roles as primary pathogens, co-infection agents, or commensals.
Free PMC Article
10.
Liu S, Zhao L, Zhai Z, Zhao W, Ding J, Dai R, Sun T, Meng H.
Curr Microbiol. 2015 Dec;71(6):643-9. doi: 10.1007/s00284-015-0895-6. Epub 2015 Aug 29.
.. A little was known regarding the status of gut microbiota after piglets infected by PEDV. In this study, aided by metagenome sequencing technology, gut microbiota profiles in feces of viral diarrhea (VD) and viral control (VC) piglets were investigated. 
The results showed that the abundance of four dominant phyla (Fusobacteria, Actinobacteria, Verrucomicrobia, and Proteobacteria) in feces was affected greatly by porcine epidemic diarrhea. Especially, the abundance of Fusobacteria was higher in VD piglets (36%) than in VC piglets (5%). On the contrary, the Verrucomicrobia was detected in lower distribution proportion in VD piglets (around 0%) than in VC piglets (20%). Furthermore, 25 genera were significantly different between VC and VD piglets at the genus level. Among the 25 genera, Leptotrichia belonging to Fusobacteria was remarkably lower in VC piglets than in VD piglets. Akkermansia belonging to Verrucomicrobia was higher in VC piglets than in VD piglets. Our findings implicated that the gut microbiota associated with PED significantly provided an insight into the pathology and physiology of PED.
11. (PED vaikutus pitemmällä ajalla: dysbioosi  heijastumaa).
Koh HW, Kim MS, Lee JS, Kim H, Park SJ.
Microbes Environ. 2015;30(3):284-7. doi: 10.1264/jsme2.ME15046. Epub 2015 Jul 25.
The gastrointestinal tract of mammals is a complex ecosystem with distinct environments and comprises hundreds of different types of bacterial cells. The gut microbiota may play a critical role in the gut health of the host. We herein attempted to identify a microbiota shift that may be affected by porcine epidemic diarrhea (PED). We observed significant differences in microbiota between the control and PED virus (PEDV)-infected groups at both the phylum and genus level. Most commensal bacteria (i.e. Psychrobacter, Prevotella, and Faecalibacterium) in the healthy gastrointestinal tract were decreased due to dysbiosis induced by PEDV infection.Free PMC Article
12.
Kuba K, Imai Y, Penninger JM.
Circ J. 2013;77(2):301-8. Epub 2013 Jan 18. Revie  Abstract
Angiotensin-converting enzyme 2 (ACE2) is a negative regulator of the renin-angiotensin system, and functions as the key SARS coronavirus receptor and stabilizer of neutral amino acid transporters. ACE2 catalyzes the conversion of angiotensin II to angiotensin 1-7, thereby counterbalancing ACE activity. Accumulating evidence indicates that the enzymatic activity of ACE2 has a protective role in cardiovascular diseases. Loss of ACE2 can be detrimental, as it leads to functional deterioration of the heart and progression of cardiac, renal, and vascular pathologies. Recombinant soluble human ACE2 protein has been demonstrated to exhibit beneficial effects in various animal models, including cardiovascular diseases. ACE2 is a multifunctional enzyme and thus potentially acts on other vasoactive peptides, such as Apelin, a vital regulator of blood pressure and myocardium contractility. In addition, ACE2 is structurally a chimeric protein that has emerged from the duplication of 2 genes: homology with ACE at the carboxypeptidase domain and homology with Collectrin in the transmembrane C-terminal domain. ACE2 has been implicated in the pathology of Hartnup's disease, a disorder of amino acid homeostasis, and, via its function in amino acid transport, it has been recently revealed that ACE2 controls intestinal inflammation and diarrhea, thus regulating the gut microbiome. This review summarizes and discusses the structure and multiple functions of ACE2 and the relevance of this key enzyme in disease pathogenesis.
Free Article

Inga kommentarer:

Skicka en kommentar