Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
J Virol ; 95(24): e0134521, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34586858

ABSTRACT

Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, causes serious diarrhea in suckling piglets and has the potential for cross-species transmission. Although extensive studies have been reported on the biology and pathogenesis of PDCoV, the mechanisms by which PDCoV enters cells are not well characterized. In this study, we investigated how PDCoV enters IPI-2I cells, a line of porcine intestinal epithelial cells derived from pig ileum. Immunofluorescence assays, small interfering RNA (siRNA) interference, specific pharmacological inhibitors, and dominant negative mutation results revealed that PDCoV entry into IPI-2I cells depended on clathrin, dynamin, and a low-pH environment but was independent of caveolae. Specific inhibition of phosphatidylinositol 3-kinase (PI3K) and the Na+/H+ exchanger (NHE) revealed that PDCoV entry involves macropinocytosis and depends on NHE rather than on PI3K. Additionally, Rab5 and Rab7, but not Rab11, regulated PDCoV endocytosis. This is the first study to demonstrate that PDCoV uses clathrin-mediated endocytosis and macropinocytosis as alternative endocytic pathways to enter porcine intestinal epithelial cells. We also discussed the entry pathways of PDCoV into other porcine cell lines. Our findings reveal the entry mechanisms of PDCoV and provide new insight into the PDCoV life cycle. IMPORTANCE An emerging enteropathogenic coronavirus, PDCoV, has the potential for cross-species transmission, attracting extensive attenuation. Characterizing the detailed process of PDCoV entry into cells will deepen our understanding of the viral infection and pathogenesis and provide clues for therapeutic intervention against PDCoV. With the objective, we used complementary approaches to dissect the process in PDCoV-infected IPI-2I cells, a line of more physiologically relevant intestinal epithelial cells to PDCoV infection in vivo. Here, we demonstrate that PDCoV enters IPI-2I cells via macropinocytosis, which does not require a specific receptor, and clathrin-mediated endocytosis, which requires a low-pH environment and dynamin, while a caveola-mediated endocytic pathway is used by PDCoV to enter swine testicular (ST) cells and porcine kidney (LLC-PK1) cells. These findings provide a molecular detail of the cellular entry pathways of PDCoV and may direct us toward novel antiviral drug development.


Subject(s)
Coronavirus Infections/virology , Deltacoronavirus/physiology , Dynamins/metabolism , Endocytosis , Epithelial Cells/virology , Animals , Cell Line , Cell Survival , Clathrin/metabolism , Coronavirus/genetics , Hydrogen-Ion Concentration , Ileum/virology , Kidney/virology , Phosphatidylinositol 3-Kinases/metabolism , Pinocytosis , RNA, Small Interfering/metabolism , Swine , Swine Diseases/virology , Virus Internalization , rab5 GTP-Binding Proteins/metabolism
2.
Virology ; 558: 119-125, 2021 06.
Article in English | MEDLINE | ID: mdl-33756424

ABSTRACT

Coinfection of porcine epidemic diarrhea virus (PEDV) and porcine deltacoronavirus (PDCoV) is one of common findings in diarrheal piglets that cause massive economic losses to the pig industry globally. However, the mechanism of the co-infection is unclear. In this study, neonatal non-colostrum-fed piglets were exposed orally with a single infection of PDCoV or PEDV, or coinfection of PDCoV and PEDV. Clinically all viral infected piglets developed watery diarrhea and dehydration in 24 h post-exposure (hpe) and were succumbed to viral diarrhea disease and euthanized at 72 hpe. Histopathologically, acute gastroenteritis is evident in all viral infected piglet. Immunohistochemistry, RNAscope and RT-qPCR demonstrated that PEDV tropism changes from epithelial cells of small intestine to gastric epithelial cells and macrophages in Peyer's patches in the ileum. These findings suggest that coinfection of PDCoV and PEDV can alter PEDV tropism that may affect the outcome of viral disease in piglets. This animal model can be used for the pathogenesis and vaccination of viral coinfection in piglet in the future.


Subject(s)
Coinfection/virology , Coronavirus Infections/veterinary , Deltacoronavirus/pathogenicity , Gastrointestinal Tract/virology , Porcine epidemic diarrhea virus/pathogenicity , Viral Tropism , Animals , Coronavirus Infections/virology , Diarrhea/virology , Disease Models, Animal , Epithelial Cells/virology , Ileum/virology , Swine
3.
Viruses ; 13(1)2021 Jan 11.
Article in English | MEDLINE | ID: mdl-33440894

ABSTRACT

Norovirus genogroup II (NoV GII) induces acute gastrointestinal food-borne illness in humans. Because gnotobiotic pigs can be infected with human norovirus (HuNoV) GII, they are frequently used to analyze the associated pathogenic mechanisms and immune responses, which remain poorly understood. Recently, mRNA sequencing analysis (RNA-Seq) has been used to identify cellular responses to viruses. In this study, we investigated the host immune response and possible mechanisms involved in virus evasion in the ileum of gnotobiotic pigs infected with HuNoV by RNA-Seq. HuNoV was detected in the feces, blood, and tissues of the jejunum, ileum, colon, mesenteric lymph node, and spleen of pigs infected with HuNoV. In analysis of mRNA sequencing, expression of anti-viral protein genes such as OAS1, MX1, and MX2 were largely decreased, whereas type I IFN was increased in pigs infected with HuNoV. In addition, expression of TNF and associated anti-inflammatory cytokine genes such as IL10 was increased in HuNoV-infected pigs. Expression of genes related to natural killer (NK) cell cytotoxicity and CD8+ T cell exhaustion was increased, whereas that of MHC class I genes was decreased. Expression profiles of selected genes were further confirmed by qRT-PCR and Western blot. These results suggest that infection with HuNoV induces NK cell-mediated cytotoxicity but suppresses type I IFN- and CD8+ T cell-mediated antiviral responses.


Subject(s)
Caliciviridae Infections/veterinary , Gastroenteritis/veterinary , Ileum/virology , Immunity , Norovirus/physiology , Swine Diseases/immunology , Swine Diseases/virology , Adaptive Immunity , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , High-Throughput Nucleotide Sequencing , Humans , Killer Cells, Natural , Models, Biological , RNA, Messenger , RNA, Viral , Swine , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
5.
J Crohns Colitis ; 15(3): 485-498, 2021 Mar 05.
Article in English | MEDLINE | ID: mdl-32915959

ABSTRACT

BACKGROUND: Patients with inflammatory bowel disease [IBD] are considered immunosuppressed, but do not seem more vulnerable for COVID-19. Nevertheless, intestinal inflammation has shown to be an important risk factor for SARS-CoV-2 infection and prognosis. Therefore, we investigated the role of intestinal inflammation on the viral intestinal entry mechanisms, including ACE2, in IBD. METHODS: We collected inflamed and uninflamed mucosal biopsies from Crohn's disease [CD] [n = 193] and ulcerative colitis [UC] [n = 158] patients, and from 51 matched non-IBD controls for RNA sequencing, differential gene expression, and co-expression analysis. Organoids from UC patients were subjected to an inflammatory mix and processed for RNA sequencing. Transmural ileal biopsies were processed for single-cell [sc] sequencing. Publicly available colonic sc-RNA sequencing data, and microarrays from tissue pre/post anti-tumour necrosis factor [TNF] therapy, were analysed. RESULTS: In inflamed CD ileum, ACE2 was significantly decreased compared with control ileum [p = 4.6E-07], whereas colonic ACE2 was higher in inflamed colon of CD/UC compared with control [p = 8.3E-03; p = 1.9E-03]. Sc-RNA sequencing confirmed this ACE2 dysregulation and exclusive epithelial ACE2 expression. Network analyses highlighted HNF4A as key regulator of ileal ACE2, and pro-inflammatory cytokines and interferon regulating factors regulated colonic ACE2. Inflammatory stimuli upregulated ACE2 in UC organoids [p = 1.7E-02], but not in non-IBD controls [p = 9.1E-01]. Anti-TNF therapy restored colonic ACE2 regulation in responders. CONCLUSIONS: Intestinal inflammation alters SARS-CoV-2 coreceptors in the intestine, with opposing dysregulations in ileum and colon. HNF4A, an IBD susceptibility gene, seems an important upstream regulator of ACE2 in ileum, whereas interferon signalling might dominate in colon.


Subject(s)
Angiotensin-Converting Enzyme 2/immunology , COVID-19 , Colitis, Ulcerative , Colon , Crohn Disease , Hepatocyte Nuclear Factor 4 , Ileum , Interferons/immunology , SARS-CoV-2/physiology , Biopsy/methods , COVID-19/immunology , COVID-19/pathology , COVID-19/physiopathology , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colitis, Ulcerative/virology , Colon/immunology , Colon/pathology , Colon/virology , Crohn Disease/immunology , Crohn Disease/pathology , Crohn Disease/virology , Cytokines/immunology , Female , Gene Expression Regulation , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/immunology , Humans , Ileum/immunology , Ileum/pathology , Ileum/virology , Male , Middle Aged , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis
6.
J Vet Sci ; 21(5): e78, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33016023

ABSTRACT

BACKGROUND: Enteritis is one of the most frequently reported symptoms in piglets infected with porcine circovirus type 2 (PCV2), but the immunopathogenesis has not been reported. OBJECTIVES: This study examined the effect of a PCV2 infection on the intestinal mucosal immune function through morphological observations and immune-related molecular detection. METHODS: Morphological changes within the ileum of piglets during a PCV2 infection were observed. The expression of the related-molecules was analyzed using a gene chip. The immunocyte subsets were analyzed by flow cytometry. The secretory immunoglobulin A (SIgA) content was analyzed by enzyme-linked immunosorbent assay. RESULTS: The PCV2 infection caused ileal villus damage, intestinal epithelial cells exfoliation, and an increase in lymphocytes in the lamina propria at 21 days post-infection. Differentially expressed genes occurred in the defense response, inflammatory response, and the complement and coagulation cascade reactions. Most of them were downregulated significantly at the induction site and upregulated at the effector site. The genes associated with SIgA production were downregulated significantly at the induction site. In contrast, the expression of the Toll-like receptor-related genes was upregulated significantly at the effector site. The frequencies of dendritic cells, B cells, and CD8⁺T cells were upregulated at the 2 sites. The SIgA content decreased significantly in the ileal mucosa. CONCLUSIONS: PCV2 infections can cause damage to the ileum that is associated with changes in immune-related gene expression, immune-related cell subsets, and SIgA production. These findings elucidated the molecular changes in the ileum after a PCV2 infection from the perspective of intestinal mucosal immunity, which provides insights into a further study for PCV2-induced enteritis.


Subject(s)
Circoviridae Infections/veterinary , Circovirus/physiology , Enteritis/veterinary , Ileum/metabolism , Intestinal Mucosa/metabolism , Swine Diseases/virology , Animals , Circoviridae Infections/virology , Enteritis/virology , Ileum/virology , Intestinal Mucosa/virology , Swine
7.
Cells ; 9(9)2020 09 12.
Article in English | MEDLINE | ID: mdl-32932592

ABSTRACT

Feline coronaviruses (FCoVs) infect both wild and domestic cat populations world-wide. FCoVs present as two main biotypes: the mild feline enteric coronavirus (FECV) and the fatal feline infectious peritonitis virus (FIPV). FIPV develops through mutations from FECV during a persistence infection. So far, the molecular mechanism of FECV-persistence and contributing factors for FIPV development may not be studied, since field FECV isolates do not grow in available cell culture models. In this work, we aimed at establishing feline ileum and colon organoids that allow the propagation of field FECVs. We have determined the best methods to isolate, culture and passage feline ileum and colon organoids. Importantly, we have demonstrated using GFP-expressing recombinant field FECV that colon organoids are able to support infection of FECV, which were unable to infect traditional feline cell culture models. These organoids in combination with recombinant FECVs can now open the door to unravel the molecular mechanisms by which FECV can persist in the gut for a longer period of time and how transition to FIPV is achieved.


Subject(s)
Coronavirus, Feline/growth & development , Feline Infectious Peritonitis/pathology , Organ Culture Techniques/veterinary , Organoids/growth & development , Animals , Cats , Cell Line , Colon/cytology , Colon/virology , Coronavirus, Feline/genetics , Female , HEK293 Cells , Humans , Ileum/cytology , Ileum/virology , Models, Biological , Organ Culture Techniques/methods , Organoids/cytology
8.
Vet Pathol ; 57(5): 642-652, 2020 09.
Article in English | MEDLINE | ID: mdl-32880235

ABSTRACT

In the small intestine, localized innate mucosal immunity is critical for intestinal homeostasis. Porcine epidemic diarrhea virus (PEDV) infection induces villus injury and impairs digestive function. Moreover, the infection might comprise localized innate mucosal immunity. This study investigated specific enterocyte subtypes and innate immune components of weaned pigs during PEDV infection. Four-week-old pigs were orally inoculated with PEDV IN19338 strain (n = 40) or sham-inoculated (n = 24). At day post inoculation (DPI) 2, 4, and 6, lysozyme expression in Paneth cells, cellular density of villous and Peyer's patch microfold (M) cells, and the expression of polymeric immunoglobulin receptor (pIgR) were assessed in the jejunum and ileum by immunohistochemistry, and interleukin (IL)-1ß and tumor necrosis factor (TNF)-α were measured in the jejunum by ELISA. PEDV infection led to a decrease in the ratios of villus height to crypt depth (VH-CD) in jejunum at DPI 2, 4, and 6 and in ileum at DPI 4. The number of villous M cells was reduced in jejunum at DPI 4 and 6 and in ileum at DPI 6, while the number of Peyer's patch M cells in ileum increased at DPI 2 and then decreased at DPI 6. PEDV-infected pigs also had reduced lysozyme expression in ileal Paneth cells at DPI 2 and increased ileal pIgR expression at DPI 4. There were no significant changes in IL-1ß and TNF-α expression in PEDV-infected pigs compared to controls. In conclusion, PEDV infection affected innate mucosal immunity of weaned pigs through alterations in Paneth cells, villous and Peyer's patch M cells, and pIgR expression.


Subject(s)
Coronavirus Infections/veterinary , Immunity, Innate , Intestinal Mucosa/immunology , Porcine epidemic diarrhea virus , Animals , Coronavirus Infections/immunology , Coronavirus Infections/pathology , Cytokines/analysis , Ileum/immunology , Ileum/pathology , Ileum/virology , Intestinal Mucosa/chemistry , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Jejunum/immunology , Jejunum/pathology , Jejunum/virology , Receptors, Polymeric Immunoglobulin/metabolism , Swine , Weaning
9.
Science ; 369(6499): 50-54, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32358202

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can cause coronavirus disease 2019 (COVID-19), an influenza-like disease that is primarily thought to infect the lungs with transmission through the respiratory route. However, clinical evidence suggests that the intestine may present another viral target organ. Indeed, the SARS-CoV-2 receptor angiotensin-converting enzyme 2 (ACE2) is highly expressed on differentiated enterocytes. In human small intestinal organoids (hSIOs), enterocytes were readily infected by SARS-CoV and SARS-CoV-2, as demonstrated by confocal and electron microscopy. Enterocytes produced infectious viral particles, whereas messenger RNA expression analysis of hSIOs revealed induction of a generic viral response program. Therefore, the intestinal epithelium supports SARS-CoV-2 replication, and hSIOs serve as an experimental model for coronavirus infection and biology.


Subject(s)
Betacoronavirus/physiology , Enterocytes/virology , Ileum/virology , Virus Replication , Angiotensin-Converting Enzyme 2 , Betacoronavirus/ultrastructure , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Cell Proliferation , Culture Media , Enterocytes/metabolism , Enterocytes/ultrastructure , Gene Expression , Humans , Ileum/metabolism , Ileum/ultrastructure , Lung/virology , Male , Organoids , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Virus/genetics , Receptors, Virus/metabolism , Respiratory Mucosa/virology , Severe acute respiratory syndrome-related coronavirus/physiology , SARS-CoV-2
11.
Cell Commun Signal ; 18(1): 23, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32046726

ABSTRACT

BACKGROUND: Porcine circovirus type 2 (PCV2) is the causative agent of postweaning multisystemic wasting syndrome, and is associated with a number of other diseases. PCV2 is widely distributed in most developed swine industries, and is a severe economic burden. With an eye to developing an effective, safe, and convenient vaccine against PCV2-associated diseases, we have constructed a recombinant Bacillus subtilis strain (B. subtilis-Cap) that expresses the PCV2 capsid protein (Cap). METHODS: Electroporation of a plasmid shuttle vector encoding the PCV2 Cap sequence was use to transform Bacillus subtilis. Flow cytometry was used to evaluate in vitro bone marrow derived dendritic cell (BM-DC) maturation and T cell proliferation induced by B. subtilis-Cap. Orally inoculated piglets were used for in vivo experiments; ELISA and western blotting were used to evaluate B. subtilis-Cap induced PCV2-specific IgA and IgG levels, as well as the secretion of cytokines and the expression of Toll-like receptor 2 (TLR2) and Toll-like receptor 9 (TLR9). RESULTS: We evaluated the immune response to B. subtilis-Cap in vitro using mouse BM-DCs and in vivo using neonatal piglets orally inoculated with B. subtilis-Cap. Our results showed that the recombinant B. subtilis-Cap activated BM-DCs, significantly increased co-stimulatory molecules (CD40 and CD80) and major histocompatibility complex II, and induced allogenic T cells proliferation. Piglets immunized with B. subtilis-Cap had elevated levels of PCV2-specific IgA in the mucosal tissues of the digestive and respiratory tract, and PCV2-specific IgG in serum (P < 0.05 or P < 0.01). Ileal immunocompetent cells, such as the IgA-secreting cells (P < 0.01), intestinal intraepithelial lymphocytes (IELs) (P < 0.01), CD3+ T lymphocytes (P < 0.01) and CD4+ T lymphocytes (P < 0.01) increased significantly in the B. subtilis-Cap immunized piglets. Additionally, B. subtilis-Cap inoculation resulted in increased the expression of TLR2 and TLR9 (P < 0.01), and induced the secretion of cytokines IL-1ß, IL-6, interferon-γ, and ß-defensin 2 (P < 0.01). CONCLUSIONS: We constructed a prototype PCV2 vaccine that can be administered orally and elicits a more robust humoral and cellular immunity than inactivated PCV2. B. subtilis-Cap is a promising vaccine candidate that is safe, convenient, and inexpensive. Further in vivo research is needed to determine its full range of efficacy in pigs.


Subject(s)
Capsid Proteins/administration & dosage , Circoviridae Infections/immunology , Circoviridae Infections/virology , Immunity , Immunization , Recombinant Proteins/administration & dosage , Administration, Oral , Animals , Bacillus subtilis , Cell Differentiation , Cell Proliferation , Circovirus , Cytokines/metabolism , Dendritic Cells/metabolism , Genetic Vectors/genetics , Ileum/immunology , Ileum/pathology , Ileum/virology , Immunoglobulin A/metabolism , Mice, Inbred C57BL , Phenotype , Plasmids/genetics , Swine , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism
12.
BMJ Case Rep ; 12(11)2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31772132

ABSTRACT

Cytomegalovirus (CMV) infection of the gastrointestinal tract is common in immunosuppressed patients; however, small bowel perforation from tissue-invasive CMV disease after many years of immunosuppressive therapy is a rare complication requiring timely medical and surgical intervention. We report a case of a postrenal transplant patient who presented to the emergency department with severe lower abdominal pain with CT of the abdomen/pelvis revealing a small bowel perforation. He underwent an emergent laparoscopic right hemicolectomy, and his histopathology of the terminal ileum was positive for CMV disease. He was successfully treated with intravenous ganciclovir postoperatively. We discuss the pathophysiology, histopathological features and treatment of CMV infection.


Subject(s)
Abdominal Pain/etiology , Cytomegalovirus Infections/complications , Intestinal Perforation/diagnostic imaging , Transplants/virology , Abdominal Pain/diagnosis , Administration, Intravenous , Aged , Antiviral Agents/administration & dosage , Antiviral Agents/therapeutic use , Colectomy/methods , Cytomegalovirus/metabolism , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/virology , Diagnosis, Differential , Ganciclovir/administration & dosage , Ganciclovir/therapeutic use , Humans , Ileum/pathology , Ileum/virology , Immunocompromised Host , Intestinal Perforation/drug therapy , Intestinal Perforation/etiology , Intestinal Perforation/surgery , Kidney Transplantation/adverse effects , Laparoscopy/methods , Male , Transplants/drug effects , Treatment Outcome
13.
J Gen Virol ; 100(11): 1530-1540, 2019 11.
Article in English | MEDLINE | ID: mdl-31596195

ABSTRACT

The role of commensal microbiota in enteric viral infections has been explored extensively, but the interaction between human gut microbiota (HGM) and human norovirus (HuNoV) is poorly understood. In this study, we established an HGM-Transplanted gnotobiotic (Gn) pig model of HuNoV infection and disease, using an infant stool as HGM transplant and a HuNoV GII.4/2006b strain for virus inoculation. Compared to germ-free Gn pigs, HuNoV inoculation in HGMT Gn pigs resulted in increased HuNoV shedding, characterized by significantly higher shedding titres on post inoculation day (PID) 3, 4, 6, 8 and 9, and significantly longer mean duration of virus shedding. In addition, virus titres were significantly higher in duodenum and distal ileum of HGMT Gn pigs on PID10, while comparable and transient HuNoV viremia was detected in both groups. 16S rRNA gene sequencing demonstrated that HuNoV infection dramatically altered intestinal microbiota in HGMT Gn pigs at the phylum (Proteobacteria, Firmicutes and Bacteroidetes) and genus (Enterococcus, Bifidobacterium, Clostridium, Ruminococcus, Anaerococcus, Bacteroides and Lactobacillus) levels. In summary, enhanced GII.4 HuNoV infection was observed in the presence of HGM, and host microbiota was susceptible to disruption upon HuNoV infection.


Subject(s)
Caliciviridae Infections/pathology , Dysbiosis , Gastrointestinal Microbiome , Microbial Interactions , Microbiota , Norovirus/growth & development , Animals , Blood/virology , Caliciviridae Infections/complications , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Disease Models, Animal , Duodenum/virology , Fecal Microbiota Transplantation , Genotype , Germ-Free Life , Humans , Ileum/virology , Norovirus/classification , Norovirus/genetics , Phylogeny , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Swine , Time Factors , Viral Load , Virus Shedding
14.
Cell ; 179(3): 644-658.e13, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31607511

ABSTRACT

Rotavirus (RV) encounters intestinal epithelial cells amidst diverse microbiota, opening possibilities of microbes influencing RV infection. Although RV clearance typically requires adaptive immunity, we unintentionally generated RV-resistant immunodeficient mice, which, we hypothesized, reflected select microbes protecting against RV. Accordingly, such RV resistance was transferred by co-housing and fecal transplant. RV-protecting microbiota were interrogated by heat, filtration, and antimicrobial agents, followed by limiting dilution transplant to germ-free mice and microbiome analysis. This approach revealed that segmented filamentous bacteria (SFB) were sufficient to protect mice against RV infection and associated diarrhea. Such protection was independent of previously defined RV-impeding factors, including interferon, IL-17, and IL-22. Colonization of the ileum by SFB induced changes in host gene expression and accelerated epithelial cell turnover. Incubation of RV with SFB-containing feces reduced infectivity in vitro, suggesting direct neutralization of RV. Thus, independent of immune cells, SFB confer protection against certain enteric viral infections and associated diarrheal disease.


Subject(s)
Adaptive Immunity/genetics , Diarrhea/microbiology , Intestinal Mucosa/microbiology , Rotavirus Infections/microbiology , Animals , Anti-Infective Agents/pharmacology , Bacteria/genetics , Bacteria/metabolism , Diarrhea/prevention & control , Diarrhea/virology , Feces/microbiology , Gene Expression Regulation/genetics , Humans , Ileum/microbiology , Ileum/pathology , Ileum/virology , Interferons/genetics , Interleukin-17/genetics , Interleukins/genetics , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Mice , Microbiota/genetics , Rotavirus/pathogenicity , Rotavirus Infections/prevention & control , Rotavirus Infections/virology , Interleukin-22
15.
Viruses ; 11(6)2019 06 05.
Article in English | MEDLINE | ID: mdl-31195631

ABSTRACT

Pseudorabies virus (PRV) variants broke out in china since 2011, causing high fever, respiratory distress, systemic neurological symptoms, and diarrhea in piglets. This study investigated the effect of intranasal PRV variant (AH02LA) infection on ileal and colonic bacterial communities and immune status in piglets. Ten piglets (free of PRV) were assigned to PRV variant and control groups (uninfected). At day 5 after inoculation, all piglets were euthanized. No PRV was detected in the ileal and colonic mucosa. In the PRV group, we observed up-regulation of specific cytokines gene expression, down-regulation of intestinal barrier-related gene expression, and reduction of secretory immunoglobulin A (sIgA) concentration in the ileum and colon. PRV infection increased the diversity of ileal bacterial community composition. PRV infection reduced the abundance of some beneficial bacteria (Lactobacillus species in the ileum and colon; butyrate-producing bacteria species in the colon) and increased the abundance of potentially pathogenic Fusobacterium nucleatum in the ileum and Sphingomonas paucimobilis in the colon. Moreover, PRV infection decreased concentrations of the beneficial lactate in the ileum and butyrate in the colon. However, this study does not allow to evaluate whether the observed changes are directly due to the PRV infection or rather to indirect effects (fever, clinical signs and changes in diet), and will be our next research content. In summary, our findings provide evidence that intranasal PRV infection directly or indirectly brings gut health risks and implications, although no PRV was detected in the ileum and colon.


Subject(s)
Colon/microbiology , Herpesvirus 1, Suid , Ileum/microbiology , Swine Diseases/virology , Administration, Intranasal , Animals , Butyrates/analysis , Colon/immunology , Colon/metabolism , Colon/virology , Cytokines/metabolism , Fusobacterium/growth & development , Ileum/immunology , Ileum/metabolism , Ileum/virology , Immunoglobulin A/metabolism , Lactic Acid/analysis , Lactobacillus/growth & development , Microbial Interactions , Microbiota , Pseudorabies/pathology , Pseudorabies/virology , Sphingomonas/growth & development , Swine
16.
Virology ; 527: 77-88, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30468938

ABSTRACT

The composition of gastrointestinal tract viromes has been associated with multiple diseases. Our understanding of virus communities in the GI tract is still very limited due to challenges in sampling from different GI sites. Here we defined the GI viromes of 15 rhesus macaques with chronic diarrhea. Luminal content samples from terminal ileum, proximal and distal colon were collected at necropsy while samples from the rectum were collected antemortem using a fecal loop. The composition of and ecological parameters associated with the terminal ileum virome were distinct from the colon and rectum samples; these differences were driven by bacteriophages rather than eukaryotic viruses. The six contigs that were most discriminative of the viromes were distantly related to bacteriophages from three different families. Our analysis provides support for using fecal loop sampling of the rectum as a proxy of the colonic virome in humans.


Subject(s)
Bacteriophages/physiology , Biodiversity , Diarrhea/veterinary , Lower Gastrointestinal Tract/virology , Macaca mulatta , Primate Diseases/virology , Animals , Bacteriophages/classification , Bacteriophages/genetics , Chronic Disease , Colon/pathology , Colon/virology , Contig Mapping , Diarrhea/virology , Feces/virology , Ileum/pathology , Ileum/virology , Lower Gastrointestinal Tract/pathology , Metagenome , Rectum/virology
17.
J Virol ; 93(5)2019 03 01.
Article in English | MEDLINE | ID: mdl-30541861

ABSTRACT

Porcine epidemic diarrhea virus (PEDV), a member of the group of alphacoronaviruses, is the pathogen of a highly contagious gastrointestinal swine disease. The elucidation of the events associated with the intestinal epithelial response to PEDV infection has been limited by the absence of good in vitro porcine intestinal models that recapitulate the multicellular complexity of the gastrointestinal tract. Here, we generated swine enteroids from the intestinal crypt stem cells of the duodenum, jejunum, or ileum and found that the generated enteroids are able to satisfactorily recapitulate the complicated intestinal epithelium in vivo and are susceptible to infection by PEDV. PEDV infected multiple types of cells, including enterocytes, stem cells, and goblet cells, and exhibited segmental infection discrepancies compared with ileal enteroids and colonoids, and this finding was verified in vivo Moreover, the clinical isolate PEDV-JMS propagated better in ileal enteroids than the cell-adapted isolate PEDV-CV777, and PEDV infection suppressed interferon (IFN) production early during the infection course. IFN lambda elicited a potent antiviral response and inhibited PEDV in enteroids more efficiently than IFN alpha (IFN-α). Therefore, swine enteroids provide a novel in vitro model for exploring the pathogenesis of PEDV and for the in vitro study of the interplay between a host and a variety of swine enteric viruses.IMPORTANCE PEDV is a highly contagious enteric coronavirus that causes significant economic losses, and the lack of a good in vitro model system is a major roadblock to an in-depth understanding of PEDV pathogenesis. Here, we generated a porcine intestinal enteroid model for PEDV infection. Utilizing porcine intestinal enteroids, we demonstrated that PEDV infects multiple lineages of the intestinal epithelium and preferably infects ileal enteroids over colonoids and that enteroids prefer to respond to IFN lambda 1 over IFN-α. These events recapitulate the events that occur in vivo This study constitutes the first use of a primary intestinal enteroid model to investigate the susceptibility of porcine enteroids to PEDV and to determine the antiviral response following infection. Our study provides important insights into the events associated with PEDV infection of the porcine intestine and provides a valuable in vitro model for studying not only PEDV but also other swine enteric viruses.


Subject(s)
Coronavirus Infections/immunology , Gastrointestinal Diseases/veterinary , Immunity, Innate/immunology , Intestinal Mucosa/immunology , Porcine epidemic diarrhea virus/immunology , Animals , Cell Line , Chlorocebus aethiops , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Duodenum/cytology , Duodenum/virology , Gastrointestinal Diseases/virology , Ileum/cytology , Ileum/virology , Interferons/biosynthesis , Intestinal Mucosa/virology , Jejunum/cytology , Jejunum/virology , Models, Biological , Porcine epidemic diarrhea virus/physiology , Swine , Swine Diseases/virology , Vero Cells
18.
Virology ; 527: 89-97, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30471453

ABSTRACT

Long noncoding RNAs (lncRNAs) have been implicated in various life processes. However, the lncRNA expression and potential functions in porcine endemic diarrhea virus (PEDV) infection and host defense are still poorly understood. In this study, we investigated the lncRNA expression profiles during PEDV infection in intestinal porcine epithelial cell-jejunum 2 (IPEC-J2) cell lines by next-generation sequencing and identified 6188 novel lncRNAs. The functional annotation analysis revealed that these lncRNAs might be associated with many immunity-related genes. We next selected candidate lncRNAs related to immune response pathways and further identified their differential expression in PEDV-infected IPEC-J2 cells and newborn piglets. Our results demonstrated that PEDV infection regulated lncRNA expression patterns in both the IPEC-J2 cell line and piglet ileum. These findings provide the first large-scale survey of lncRNAs associated with PEDV infection, specifically the lncRNAs responsible for the activation of the immune system within the ileum.


Subject(s)
Coronavirus Infections/veterinary , Gene Expression Regulation , Porcine epidemic diarrhea virus/physiology , RNA, Long Noncoding/genetics , Swine Diseases/virology , Animals , Cell Line , Coronavirus Infections/genetics , Coronavirus Infections/immunology , Coronavirus Infections/virology , Gene Expression Profiling , Host-Pathogen Interactions , Ileum/immunology , Ileum/virology , Swine , Swine Diseases/genetics , Swine Diseases/immunology
19.
In Vivo ; 33(1): 251-254, 2019.
Article in English | MEDLINE | ID: mdl-30587632

ABSTRACT

Pyogenic granuloma (PG) represents a lobular capillary proliferation commonly seen in the skin or oral mucosa. They are rarely reported in the gastrointestinal tract. The mechanism underlying PG pathogenesis is not well understood. Only one case of cutaneous PG associated with cytomegalovirus (CMV) infection has been reported in the English literature. Here, we report such a unique case of PG arising from the small bowel. A 67-year-old male, status post ileocolic resection, presented for follow-up colonoscopy because of Crohn's disease of the terminal ileum and the colon. Colonoscopy revealed inflammation at the ileocolic anastomosis as well as an 8-mm pedunculated lesion with an irregular surface in the neo-terminal ileum. Histological studies of the small bowel mucosa revealed chronic active ileitis with pyloric gland metaplasia, consistent with his clinical history of Crohn's disease. The lesion demonstrated a lobular architecture consisting of clusters of small capillaries of various sizes lined by a single layer of cytologically bland endothelial cells, and accompanied by acute and chronic inflammatory infiltrates and surface erosion/ulceration. The histological features supported the diagnosis of PG. Scattered viral inclusions with positive CMV immunoreactivity were present in the endothelial cells and glandular cells of pyloric gland metaplasia within the PG. To the best of our knowledge, this is the first documented case of PG with local CMV infection in patients with inflammatory bowel disease.


Subject(s)
Crohn Disease/pathology , Cytomegalovirus Infections/pathology , Granuloma, Pyogenic/pathology , Inflammatory Bowel Diseases/pathology , Aged , Crohn Disease/complications , Crohn Disease/diagnosis , Crohn Disease/virology , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/virology , Gastrointestinal Tract/pathology , Gastrointestinal Tract/virology , Granuloma, Pyogenic/complications , Granuloma, Pyogenic/diagnosis , Granuloma, Pyogenic/virology , Humans , Ileum/pathology , Ileum/virology , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/virology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Intestine, Small/pathology , Intestine, Small/virology , Male
20.
Clin Infect Dis ; 69(8): 1320-1328, 2019 09 27.
Article in English | MEDLINE | ID: mdl-30590412

ABSTRACT

BACKGROUND: Optimization of combination antiretroviral therapy (cART) can impact the human immunodeficiency virus (HIV) reservoir. We evaluated the effect on the HIV reservoir in peripheral blood and ileum biopsies in patients switching from boosted protease inhibitor (PI/r)-based therapy to dolutegravir (DTG)-based therapy. METHODS: Impact of Integrase-inhibitor DOlutegravir On the viral Reservoir (INDOOR) is a phase 4 open-label clinical trial that randomly included 42 HIV type 1-infected individuals on effective cART: 20 who switched from PI/r-based to DTG-based cART (switch group), and 22 who remained in PI/r-based regimens (control group). We analyzed blood and ileum biopsies to quantify episomal, total, and integrated HIV DNA, cell-associated HIV RNA, residual plasma viremia, T-cell subsets, cell activation, and inflammation markers. RESULTS: There were no related adverse events or treatment discontinuations due to drug intolerance. The HIV reservoir was consistently larger in ileal than in peripheral CD4+ T cells in both groups (P < .01). Residual viremia in plasma decreased in the switch group (P = .03). However, we did not observe significant longitudinal changes in low-level viral replication, total and integrated HIV reservoir, HIV transcription, T-cell maturation subsets, immunoactivation markers, inflammatory soluble proteins, or cellular markers of latently infected cells. CONCLUSIONS: The INDOOR study is the first evaluation of changes in HIV reservoir size in ileum biopsies and in peripheral blood in individuals switched from PI/r- to DTG-based cART. Although this switch was safe and well tolerated, it had no impact on a large array of immunological and inflammatory markers or on HIV reservoir markers in peripheral or in ileal CD4+ T cells. CLINICAL TRIALS REGISTRATION: EudraCT 2014-004331-39.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/drug therapy , HIV Integrase Inhibitors/therapeutic use , HIV Protease Inhibitors/therapeutic use , HIV/drug effects , Heterocyclic Compounds, 3-Ring/therapeutic use , Biopsy , Female , HIV/physiology , HIV Infections/virology , Humans , Ileum/virology , Male , Middle Aged , Oxazines , Piperazines , Pyridones , Viremia/drug therapy , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...