Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 882
Filter
1.
Virulence ; 15(1): 2360133, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38803081

ABSTRACT

Norovirus (NV) infection causes acute gastroenteritis in children and adults. Upon infection with NV, specific CD8+ T cells, which play an important role in anti-infective immunity, are activated in the host. Owing to the NV's wide genotypic variability, it is challenging to develop vaccines with cross-protective abilities against infection. To aid effective vaccine development, we examined specific CD8+ T-cell responses towards viral-structural protein (VP) epitopes, which enable binding to host susceptibility receptors. We isolated peripheral blood mononuclear cells from 196 participants to screen and identify predominant core peptides towards NV main and small envelope proteins using ex vivo and in vitro intracellular cytokine staining assays. Human leukocyte antigen (HLA) restriction characteristics were detected using next-generation sequencing. Three conservative immunodominant VP-derived CD8+ T-cell epitopes, VP294-102 (TDAARGAIN), VP2153-161 (RGPSNKSSN), and VP1141-148 (FPHIIVDV), were identified and restrictively presented by HLA-Cw * 0102, HLA-Cw * 0702, and HLA-A *1101 alleles, separately. Our findings provide useful insights into the development of future vaccines and treatments for NV infection.


Subject(s)
CD8-Positive T-Lymphocytes , Caliciviridae Infections , Capsid Proteins , Epitopes, T-Lymphocyte , Gastroenteritis , Norovirus , Humans , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/immunology , Capsid Proteins/genetics , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Norovirus/immunology , Norovirus/genetics , Adult , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/genetics , Male , Gastroenteritis/virology , Gastroenteritis/immunology , Female , Middle Aged , Young Adult , Child , Adolescent , Leukocytes, Mononuclear/immunology , Immunodominant Epitopes/immunology , Child, Preschool , Aged
2.
N Engl J Med ; 389(19): 1790-1796, 2023 Nov 09.
Article in English | MEDLINE | ID: mdl-37937778

ABSTRACT

Immune checkpoint blockade has become standard treatment for many types of cancer. Such therapy is indicated most often in patients with advanced or metastatic disease but has been increasingly used as adjuvant therapy in those with early-stage disease. Adverse events include immune-related organ inflammation resembling autoimmune diseases. We describe a case of severe immune-related gastroenterocolitis in a 4-month-old infant who presented with intractable diarrhea and failure to thrive after in utero exposure to pembrolizumab. Known causes of the symptoms were ruled out, and the diagnosis of pembrolizumab-induced immune-related gastroenterocolitis was supported by the results of histopathological assays, immunophenotyping, and analysis of the level of antibodies against programmed cell death protein 1 (PD-1). The infant's condition was successfully treated with prednisolone and infliximab.


Subject(s)
Gastroenteritis , Immune Checkpoint Inhibitors , Neoplasms , Humans , Infant , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Enteritis/chemically induced , Enteritis/diagnosis , Enteritis/drug therapy , Enteritis/immunology , Neoplasms/drug therapy , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Failure to Thrive/chemically induced , Failure to Thrive/immunology , Diarrhea, Infantile/chemically induced , Diarrhea, Infantile/immunology , Gastroenteritis/chemically induced , Gastroenteritis/diagnosis , Gastroenteritis/drug therapy , Gastroenteritis/immunology , Enterocolitis/chemically induced , Enterocolitis/diagnosis , Enterocolitis/drug therapy , Enterocolitis/immunology , Programmed Cell Death 1 Receptor/immunology
3.
Gut Microbes ; 15(2): 2249960, 2023 12.
Article in English | MEDLINE | ID: mdl-37655966

ABSTRACT

Over 90% of epidemic non-bacterial gastroenteritis are caused by human noroviruses (NoVs), which persist in a substantial subset of people allowing their spread worldwide. This has led to a significant number of endemic cases and up to 70,000 children deaths in developing countries. NoVs are primarily transmitted through the fecal-oral route. To date, studies have focused on the influence of the gut microbiota on enteric viral clearance by mucosal immunity. In this study, the use of mouse norovirus S99 (MNoV_S99) and CR6 (MNoV_CR6), two persistent strains, allowed us to provide evidence that the norovirus-induced exacerbation of colitis severity relied on bacterial sensing by nucleotide-binding oligomerization domain 2 (Nod2). Consequently, Nod2-deficient mice showed reduced levels of gravity of Dextran sodium sulfate (DSS)-induced colitis with both viral strains. And MNoV_CR6 viremia was heightened in Nod2-/- mice in comparison with animals hypomorphic for Atg16l1, which are prone to aggravated inflammation under DSS. Accordingly, the infection of macrophages derived from WT mice promoted the phosphorylation of Signal Transducer and Activator of Transcription 1 (STAT1) and NOD2's expression levels. Higher secretion of Tumor Necrosis Factor alpha (TNFα) following NOD2 activation and better viral clearance were measured in these cells. By contrast, reduced levels of pSTAT1 and blunted downstream secretion of TNFα were found in Nod2-deficient macrophages infected by MNoV_S99. Hence, our results uncover a previously unidentified virus-host-bacterial interplay that may represent a novel therapeutic target for treating noroviral origin gastroenteritis that may be linked with susceptibility to several common illnesses such as Crohn's disease.


Subject(s)
Caliciviridae Infections , Colitis , Gastroenteritis , Gastrointestinal Microbiome , Nod2 Signaling Adaptor Protein , Animals , Mice , Caliciviridae Infections/immunology , Colitis/chemically induced , Colitis/virology , Gastroenteritis/immunology , Gastroenteritis/virology , Nod2 Signaling Adaptor Protein/metabolism
4.
Int Immunopharmacol ; 113(Pt A): 109386, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36461593

ABSTRACT

IL-17D is a new member of the IL-17 family. Currently, it is believed that IL-17D can directly act on immune cells or may indirectly modulate immune responses by regulating cytokine expression. Herein, we hypothesized that IL-17D regulates the expression of chemokines in intestinal epithelial cells, in turn modulating the immune response within intestinal mucosa under hyperoxia. To explore this notion, newborn rats were divided into a hyperoxia group (85 % O2) and control group (21 % O2). Small intestinal tissues were obtained from neonatal rats at 3, 7, 10, and 14 days. Similarly, intestinal epithelial cells were treated by hyperoxia (85 % O2) as the hyperoxia group or were incubated under normal oxygen (21 % O2) as the control group. Finally, intestinal epithelial cells subjected to hyperoxia were treated with recombinant IL-17D and IL-17D antibodies for 24, 48, and 72 h. Immunohistochemistry, western blot, and reverse transcription-quantitative polymerase chain reaction were used to detect the expression levels of chemokines and chemokine receptors in intestinal tissues of newborn rats and intestinal epithelial cells. We found that hyperoxia affected chemokine expression both in vivo and in vitro. Under hyperoxia, IL-17D promoted the expression of CCL2, CCL25, CCL28, and CCR9 in intestinal epithelial cells while downregulating CCR2, CCR5, CCL5, and CCL20. Our findings provide a basis for further study on the effects of hyperoxia-induced intestinal inflammation and intestinal injury.


Subject(s)
Gastroenteritis , Hyperoxia , Interleukin-27 , Intestinal Mucosa , Oxygen , Animals , Rats , Chemokines/immunology , Epithelial Cells/immunology , Gastroenteritis/etiology , Gastroenteritis/immunology , Hyperoxia/complications , Hyperoxia/immunology , Immunologic Factors , Interleukin-27/immunology , Intestinal Mucosa/immunology , Intestines/immunology , Oxygen/toxicity , Receptors, Chemokine/immunology
5.
Sci Rep ; 12(1): 14275, 2022 08 22.
Article in English | MEDLINE | ID: mdl-35995986

ABSTRACT

Norovirus is a leading cause of epidemic acute gastroenteritis. More than 30 genotypes circulate in humans, some are common, and others are only sporadically detected. Here, we investigated whether serology can be used to determine which genotypes infect children. We established a multiplex protein microarray with structural and non-structural norovirus antigens that allowed simultaneous antibody testing against 30 human GI and GII genotypes. Antibody responses of sera obtained from 287 children aged < 1 month to 5.5 years were profiled. Most specific IgG and IgA responses were directed against the GII.2, GII.3, GII.4, and GII.6 capsid genotypes. While we detected antibody responses against rare genotypes, we found no evidence for wide circulation. We also detected genotype-specific antibodies against the non-structural proteins p48 and p22 in sera of older children. In this study, we show the age-dependent antibody responses to a broad range of norovirus capsid and polymerase genotypes, which will aid in the development of vaccines.


Subject(s)
Caliciviridae Infections , Gastroenteritis , Immunity, Humoral , Norovirus , Caliciviridae Infections/immunology , Capsid Proteins/genetics , Child, Preschool , Europe , Gastroenteritis/immunology , Gastroenteritis/virology , Genotype , Humans , Infant , Norovirus/genetics , Phylogeny
6.
Int J Mol Sci ; 23(3)2022 Feb 06.
Article in English | MEDLINE | ID: mdl-35163778

ABSTRACT

Innate lymphoid cells (ILCs) are a population of lymphoid cells that do not express T cell or B cell antigen-specific receptors. They are largely tissue-resident and enriched at mucosal sites to play a protective role against pathogens. ILCs mimic the functions of CD4 T helper (Th) subsets. Type 1 innate lymphoid cells (ILC1s) are defined by the expression of signature cytokine IFN-γ and the master transcription factor T-bet, involving in the type 1 immune response; ILC2s are characterized by the expression of signature cytokine IL-5/IL-13 and the master transcription factor GATA3, participating in the type 2 immune response; ILC3s are RORγt-expressing cells and are capable of producing IL-22 and IL-17 to maintain intestinal homeostasis. The discovery and investigation of ILCs over the past decades extends our knowledge beyond classical adaptive and innate immunology. In this review, we will focus on the roles of ILCs in intestinal inflammation and related disorders.


Subject(s)
Gastroenteritis/immunology , Lymphocytes/immunology , Animals , Gene Expression Regulation , Humans , Immunity, Innate , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , T-Box Domain Proteins/metabolism , Interleukin-22
7.
J Gen Virol ; 103(1)2022 01.
Article in English | MEDLINE | ID: mdl-35077345

ABSTRACT

Norovirus is the leading cause of epidemic and endemic acute gastroenteritis worldwide and the most frequent cause of foodborne illness in the United States. There is no specific treatment for norovirus infections and therapeutic interventions are based on alleviating symptoms and limiting viral transmission. The immune response to norovirus is not completely understood and mechanistic studies have been hindered by lack of a robust cell culture system. In recent years, the human intestinal enteroid/human intestinal organoid system (HIE/HIO) has enabled successful human norovirus replication. Cells derived from HIE have also successfully been subjected to genetic manipulation using viral vectors as well as CRISPR/Cas9 technology, thereby allowing studies to identify antiviral signaling pathways important in controlling norovirus infection. RNA sequencing using HIE cells has been used to investigate the transcriptional landscape during norovirus infection and to identify antiviral genes important in infection. Other cell culture platforms such as the microfluidics-based gut-on-chip technology in combination with the HIE/HIO system also have the potential to address fundamental questions on innate immunity to human norovirus. In this review, we highlight the recent advances in understanding the innate immune response to human norovirus infections in the HIE system, including the application of advanced molecular technologies that have become available in recent years such as the CRISPR/Cas9 and RNA sequencing, as well as the potential application of single cell transcriptomics, viral proteomics, and gut-on-a-chip technology to further elucidate innate immunity to norovirus.


Subject(s)
Caliciviridae Infections/immunology , Gastroenteritis/immunology , Intestines/virology , Organoids/immunology , Gastroenteritis/virology , Humans , Immunity, Innate , Intestines/immunology , Models, Biological , Norovirus/pathogenicity , Norovirus/physiology , Organoids/virology , Sequence Analysis, RNA , Virus Replication
8.
Viruses ; 13(11)2021 10 26.
Article in English | MEDLINE | ID: mdl-34834968

ABSTRACT

Noroviruses are responsible for almost a fifth of all cases of gastroenteritis worldwide. The calicivirus capsid is composed of 180 copies of VP1 with a molecular weight of ~58 kDa. This coat protein is divided into the N-terminus (N), the shell (S) and C-terminal protruding (P) domains. The S domain forms a shell around the viral RNA genome, while the P domains dimerize to form protrusions on the capsid surface. The P domain is subdivided into P1 and P2 subdomains, with the latter containing the binding sites for cellular receptors and neutralizing antibodies. Reviewed here are studies on murine norovirus (MNV) showing that the capsid responds to several physiologically relevant cues; bile, pH, Mg2+, and Ca2+. In the initial site of infection, the intestinal tract, high bile and metal concentrations and low pH cause two significant conformational changes: (1) the P domain contracts onto the shell domain and (2) several conformational changes within the P domain lead to enhanced receptor binding while blocking antibody neutralization. In contrast, the pH is neutral, and the concentrations of bile and metals are low in the serum. Under these conditions, the loops at the tip of the P domain are in the open conformation with the P domain floating on a linker or tether above the shell. This conformational state favors antibody binding but reduces interactions with the receptor. In this way, MNV uses metabolites and environmental cues in the intestine to optimize cellular attachment and escape antibody binding but presents a wholly different structure to the immune system in the serum. To our knowledge, this is the first example of a virus shapeshifting in this manner to escape the immune response.


Subject(s)
Gastroenteritis/immunology , Gastroenteritis/virology , Norovirus/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Bile , Binding Sites , Capsid/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/immunology , Cryoelectron Microscopy , Genome, Viral , Mice , Models, Molecular , Norovirus/genetics , Protein Binding , Protein Domains
9.
Viruses ; 13(10)2021 10 09.
Article in English | MEDLINE | ID: mdl-34696463

ABSTRACT

Acute gastroenteritis (AGE) is a major cause of morbidity and mortality worldwide, resulting in an estimated 440,571 deaths of children under age 5 annually. Rotavirus, norovirus, and sapovirus are leading causes of childhood AGE. A successful rotavirus vaccine has reduced rotavirus hospitalizations by more than 50%. Using rotavirus as a guide, elucidating the determinants, breath, and duration of serological antibody immunity to AGE viruses, as well as host genetic factors that define susceptibility is essential for informing development of future vaccines and improving current vaccine candidates. Here, we summarize the current knowledge of disease burden and serological antibody immunity following natural infection to inform further vaccine development for these three high-burden viruses.


Subject(s)
Gastrointestinal Diseases/immunology , Gastrointestinal Diseases/virology , Immunity, Humoral , Antibodies, Viral , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Child , Child, Preschool , Diarrhea/immunology , Diarrhea/virology , Gastroenteritis/immunology , Gastroenteritis/virology , Gastrointestinal Diseases/prevention & control , Hospitalization , Humans , Norovirus , Rotavirus , Rotavirus Infections/immunology , Rotavirus Infections/virology , Rotavirus Vaccines , Sapovirus , Vaccine Development
10.
PLoS Pathog ; 17(8): e1009719, 2021 08.
Article in English | MEDLINE | ID: mdl-34352037

ABSTRACT

Reducing food intake is a common host response to infection, yet it remains unclear whether fasting is detrimental or beneficial to an infected host. Despite the gastrointestinal tract being the primary site of nutrient uptake and a common route for infection, studies have yet to examine how fasting alters the host's response to an enteric infection. To test this, mice were fasted before and during oral infection with the invasive bacterium Salmonella enterica serovar Typhimurium. Fasting dramatically interrupted infection and subsequent gastroenteritis by suppressing Salmonella's SPI-1 virulence program, preventing invasion of the gut epithelium. Virulence suppression depended on the gut microbiota, as Salmonella's invasion of the epithelium proceeded in fasting gnotobiotic mice. Despite Salmonella's restored virulence within the intestines of gnotobiotic mice, fasting downregulated pro-inflammatory signaling, greatly reducing intestinal pathology. Our study highlights how food intake controls the complex relationship between host, pathogen and gut microbiota during an enteric infection.


Subject(s)
Bacteria/growth & development , Fasting , Gastroenteritis/prevention & control , Inflammation/prevention & control , Intestines/immunology , NF-kappa B/antagonists & inhibitors , Salmonella Infections, Animal/immunology , Salmonella typhimurium/physiology , Animals , Bacteria/immunology , Bacteria/metabolism , Female , Gastroenteritis/immunology , Gastroenteritis/microbiology , Gastrointestinal Microbiome , Inflammation/immunology , Inflammation/microbiology , Intestines/microbiology , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/complications , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology
12.
Infect Immun ; 89(10): e0008721, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34310885

ABSTRACT

Salmonella Typhimurium is a common cause of foodborne gastroenteritis and a less frequent but important cause of invasive disease, especially in developing countries. In our previous work, we showed that a live-attenuated S. Typhimurium vaccine (CVD 1921) was safe and immunogenic in rhesus macaques, although shed for an unacceptably long period (10 days) postimmunization. Consequently, we engineered a new strain, CVD 1926, which was shown to be safe and immunogenic in mice, as well as less reactogenic in mice and human cell-derived organoids than CVD 1921. In this study, we assessed the reactogenicity and efficacy of CVD 1926 in rhesus macaques. Animals were given two doses of either CVD 1926 or saline perorally. The vaccine was well-tolerated, with shedding in stool limited to a mean of 5 days. All CVD 1926-immunized animals had both a serological and a T cell response to vaccination. At 4 weeks postimmunization, animals were challenged with wild-type S. Typhimurium I77. Unvaccinated (saline) animals had severe diarrhea, with two animals succumbing to infection. Animals receiving CVD 1926 were largely protected, with only one animal having moderate diarrhea. Vaccine efficacy in this gastroenteritis model was 80%. S. Typhimurium vaccine strain CVD 1926 was safe and effective in rhesus macaques and shed for a shorter period than other previously tested live-attenuated vaccine strains. This strain could be combined with other live-attenuated Salmonella vaccine strains to create a pan-Salmonella vaccine.


Subject(s)
Gastroenteritis/immunology , Immunogenicity, Vaccine/immunology , Macaca mulatta/immunology , Salmonella Infections, Animal/immunology , Salmonella Vaccines/immunology , Salmonella typhimurium/immunology , Administration, Oral , Animals , Antibodies, Bacterial/immunology , Bacterial Proteins/immunology , Cells, Cultured , Disease Models, Animal , Female , Leukocytes, Mononuclear/immunology , Vaccination/methods
13.
Infect Immun ; 89(8): e0012121, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34097470

ABSTRACT

Salmonella enterica Typhimurium is a rod-shaped Gram-negative bacterium that mostly enters the human body through contaminated food. It causes a gastrointestinal disorder called salmonellosis in humans and typhoid-like systemic disease in mice. OmpV, an outer membrane protein of S. Typhimurium, helps in adhesion and invasion of bacteria to intestinal epithelial cells and thus plays a vital role in the pathogenesis of S. Typhimurium. In this study, we have shown that intraperitoneal immunization with OmpV is able to induce high IgG production and protection against systemic disease. Further, oral immunization with OmpV-incorporated proteoliposome (OmpV-proteoliposome [PL]) induces production of high IgA antibody levels and protection against gastrointestinal infection. Furthermore, we have shown that OmpV induces Th1 bias in systemic immunization with purified OmpV, but both Th1 and Th2 polarization in oral immunization with OmpV-proteoliposome (PL). Additionally, we have shown that OmpV activates innate immune cells, such as monocytes, macrophages, and intestinal epithelial cells, in a Toll-like receptor 2 (TLR2)-dependent manner. Interestingly, OmpV is recognized by the TLR1/2 heterodimer in monocytes, but by both TLR1/2 and TLR2/6 heterodimers in macrophages and intestinal epithelial cells. Further, downstream signaling involves MyD88, interleukin-1 receptor-associated kinase (IRAK)-1, mitogen-activated protein kinase (MAPK) (both p38 and Jun N-terminal protein kinase (JNK)), and transcription factors NF-κB and AP-1. Due to its ability to efficiently activate both the innate and adaptive immune systems and protective efficacy, OmpV can be a potential vaccine candidate against S. Typhimurium infection. Further, the fact that OmpV can be recognized by both TLR1/2 and TLR2/6 heterodimers increases its potential to act as good adjuvant in other vaccine formulations.


Subject(s)
Adhesins, Bacterial/immunology , Antigens, Bacterial/immunology , Gastroenteritis/immunology , Gastroenteritis/microbiology , Immunity , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/immunology , Animals , Biomarkers , Disease Models, Animal , Disease Susceptibility , Host-Pathogen Interactions/immunology , Mice , Signal Transduction
14.
Front Immunol ; 12: 668654, 2021.
Article in English | MEDLINE | ID: mdl-34054838

ABSTRACT

Chronic low-grade inflammation is a hallmark of obesity and associated with cardiovascular complications. However, it remains unclear where this inflammation starts. As the gut is constantly exposed to food, gut microbiota, and metabolites, we hypothesized that mucosal immunity triggers an innate inflammatory response in obesity. We characterized five distinct macrophage subpopulations (P1-P5) along the gastrointestinal tract and blood monocyte subpopulations (classical, non-classical, intermediate), which replenish intestinal macrophages, in non-obese (BMI<27kg/m2) and obese individuals (BMI>32kg/m2). To elucidate factors that potentially trigger gut inflammation, we correlated these subpopulations with cardiovascular risk factors and lifestyle behaviors. In obese individuals, we found higher pro-inflammatory macrophages in the stomach, duodenum, and colon. Intermediate blood monocytes were also increased in obesity, suggesting enhanced recruitment to the gut. We identified unhealthy lifestyle habits as potential triggers of gut and systemic inflammation (i.e., low vegetable intake, high processed meat consumption, sedentary lifestyle). Cardiovascular risk factors other than body weight did not affect the innate immune response. Thus, obesity in humans is characterized by gut inflammation as shown by accumulation of pro-inflammatory intestinal macrophages, potentially via recruited blood monocytes. Understanding gut innate immunity in human obesity might open up new targets for immune-modulatory treatments in metabolic disease.


Subject(s)
Gastroenteritis/immunology , Immunity, Innate , Immunity, Mucosal , Intestines/immunology , Macrophages/immunology , Obesity/immunology , Body Mass Index , Case-Control Studies , Diet/adverse effects , Female , Gastroenteritis/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages/metabolism , Male , Middle Aged , Obesity/complications , Obesity/metabolism , Phenotype , Prospective Studies , Risk Assessment , Risk Factors , Sedentary Behavior
15.
Front Immunol ; 12: 676628, 2021.
Article in English | MEDLINE | ID: mdl-34054868

ABSTRACT

For poultry producers, chronic low-grade intestinal inflammation has a negative impact on productivity by impairing nutrient absorption and allocation of nutrients for growth. Understanding the triggers of chronic intestinal inflammation and developing a non-invasive measurement is crucial to managing gut health in poultry. In this study, we developed two novel models of low-grade chronic intestinal inflammation in broiler chickens: a chemical model using dextran sodium sulfate (DSS) and a dietary model using a high non-starch polysaccharide diet (NSP). Further, we evaluated the potential of several proteins as biomarkers of gut inflammation. For these experiments, the chemical induction of inflammation consisted of two 5-day cycles of oral gavage of either 0.25mg DSS/ml or 0.35mg DSS/ml; whereas the NSP diet (30% rice bran) was fed throughout the experiment. At four times (14, 22, 28 and 36-d post-hatch), necropsies were performed to collect intestinal samples for histology, and feces and serum for biomarkers quantification. Neither DSS nor NSP treatments affected feed intake or livability. NSP-fed birds exhibited intestinal inflammation through 14-d, which stabilized by 36-d. On the other hand, the cyclic DSS-treatment produced inflammation throughout the entire experimental period. Histological examination of the intestine revealed that the inflammation induced by both models exhibited similar spatial and temporal patterns with the duodenum and jejunum affected early (at 14-d) whereas the ileum was compromised by 28-d. Calprotectin (CALP) was the only serum protein found to be increased due to inflammation. However, fecal CALP and Lipocalin-2 (LCN-2) concentrations were significantly greater in the induced inflammation groups at 28-d. This experiment demonstrated for the first time, two in vivo models of chronic gut inflammation in chickens, a DSS and a nutritional NSP protocols. Based on these models we observed that intestinal inflammation begins in the upper segments of small intestine and moved to the lower region over time. In the searching for a fecal biomarker for intestinal inflammation, LCN-2 showed promising results. More importantly, calprotectin has a great potential as a novel biomarker for poultry measured both in serum and feces.


Subject(s)
Dextran Sulfate/adverse effects , Diet, Carbohydrate Loading/adverse effects , Diet, Carbohydrate Loading/veterinary , Gastroenteritis/blood , Gastroenteritis/chemically induced , Poultry Diseases/blood , Poultry Diseases/chemically induced , Animal Feed , Animals , Biomarkers/metabolism , Chickens , Chronic Disease , Dextran Sulfate/administration & dosage , Dietary Fiber/adverse effects , Disease Models, Animal , Feces/chemistry , Gastroenteritis/immunology , Intestinal Mucosa/immunology , Leukocyte L1 Antigen Complex/metabolism , Lipocalin-2/metabolism , Male , Oryza/adverse effects , Poultry Diseases/immunology
16.
Mucosal Immunol ; 14(3): 751-761, 2021 05.
Article in English | MEDLINE | ID: mdl-33674763

ABSTRACT

Although they globally cause viral gastroenteritis in children, astroviruses are understudied due to the lack of well-defined animal models. While murine astroviruses (muAstVs) chronically infect immunodeficient mice, a culture system and understanding of their pathogenesis is lacking. Here, we describe a platform to cultivate muAstV using air-liquid interface (ALI) cultures derived from mouse enteroids, which support apical infection and release. Chronic muAstV infection occurs predominantly in the small intestine and correlates with higher interferon-lambda (IFN-λ) expression. MuAstV stimulates IFN-λ production in ALI, recapitulating our in vivo findings. We demonstrate that goblet cells and enterocytes are targets for chronic muAstV infection in vivo, and that infection is enhanced by parasite co-infection or type 2 cytokine signaling. Depletion of goblet cells from ALI limits muAstV infection in vitro. During chronic infection, muAstV stimulates IFN-λ production in infected cells and induces ISGs throughout the intestinal epithelium in an IFN-λ-receptor-dependent manner. Collectively, our study provides insights into the cellular tropism and innate immune responses to muAstV and establishes an enteroid-based culture system to propagate muAstV in vitro.


Subject(s)
Astroviridae Infections/immunology , Astroviridae/physiology , Cytokines/metabolism , Enterocytes/virology , Gastroenteritis/immunology , Goblet Cells/virology , Th2 Cells/immunology , Animals , Cells, Cultured , Coinfection , Enterocytes/immunology , Goblet Cells/immunology , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Viral Tropism
17.
Sci Rep ; 11(1): 6284, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737711

ABSTRACT

We aimed to investigate the endoscopic features and clinical course of CMV gastroenterocolitis in immunocompetent patients. We reviewed the medical records and endoscopic images of 86 immunocompetent patients with CMV gastroenterocolitis. Immunocompetent patients were defined as those without congenital or acquired immunodeficiency syndrome, use of anti-cancer chemotherapeutic and immunosuppressive agents, and inflammatory bowel diseases. The mean age was 65.5 ± 11.8 years and 53 (61.6%) were male. Sixty-eight (79.1%) patients had comorbidities. Upper gastrointestinal-dominant, small bowel-dominant, and colon-dominant types were observed in 19, 7, and 60 patients, respectively. Endoscopic features could be classified into discrete ulcerative type with/without exudate and diffuse erythematous type with/without exudate. Antiviral treatment with ganciclovir was initiated in 51 patients (59.3%), 40 of whom improved and 1 improved after changing ganciclovir to foscarnet. Thirty-three patients (38.4%) improved without antiviral treatment. Surgery was necessary in two patients because of colon perforation before antiviral treatment. Another two patients underwent surgery because of sigmoid stricture and cecal perforation during antiviral treatment. Endoscopic type was not associated with clinical outcomes, such as surgery and death. CMV gastroenterocolitis in immunocompetent patients mostly occur in older patients with comorbidities, and the endoscopic features vary with no association with clinical outcomes.


Subject(s)
Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/immunology , Cytomegalovirus/genetics , Cytomegalovirus/metabolism , Endoscopy, Gastrointestinal/methods , Gastroenteritis/diagnosis , Gastroenteritis/immunology , Immunocompromised Host , Adult , Aged , Aged, 80 and over , Antiviral Agents/therapeutic use , Comorbidity , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Female , Ganciclovir/therapeutic use , Gastroenteritis/drug therapy , Gastroenteritis/virology , Humans , Immunohistochemistry/methods , Male , Middle Aged , Polymerase Chain Reaction/methods , Retrospective Studies , Risk Factors , Treatment Outcome , Young Adult
18.
BMJ Case Rep ; 14(1)2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33462001

ABSTRACT

A 75-year-old man was admitted with a 3-month history of worsening diarrhoea and weight loss. He was on long-term immunosuppression following cardiac transplantation. Investigations revealed herpes simplex oesophagitis and stool samples were positive for norovirus. Treatment with acyclovir and nitazoxanide resulted in a complete resolution of symptoms. Norovirus is a common cause of infectious gastroenteritis, but immunosuppressed patients may present with chronic diarrhoea rather than an acute illness. This case highlights the importance of a low clinical threshold for testing for norovirus infection in immunocompromised patients.


Subject(s)
Caliciviridae Infections/immunology , Diarrhea/virology , Gastroenteritis/immunology , Immunocompromised Host , Norovirus/isolation & purification , Postoperative Complications/immunology , Weight Loss , Aged , Caliciviridae Infections/complications , Caliciviridae Infections/diagnosis , Chronic Disease , Diarrhea/immunology , Gastroenteritis/complications , Gastroenteritis/diagnosis , Heart Transplantation , Humans , Male , Postoperative Complications/diagnosis , Weight Loss/immunology
19.
mBio ; 11(6)2020 11 03.
Article in English | MEDLINE | ID: mdl-33144378

ABSTRACT

Chlamydia spp. productively infect mucosal epithelial cells of multiple anatomical sites, including the conjunctiva, lungs, gastrointestinal (GI) tract, and urogenital tract. We, and others, previously established that chlamydial GI tropism is mediated by distinct chromosomal and plasmid factors. In this study, we describe a genital infection-attenuated Chlamydia muridarum mutant (GIAM-1) that is profoundly and specifically attenuated in the murine genital tract. GIAM-1 infected the murine GI tract similarly to wild-type (WT) Chlamydia muridarum but did not productively infect the lower genital tract of female mice, ascend to infect the upper genital tract, or cause hydrosalpinx. However, GI infection of mice with GIAM-1 elicited a transmucosal immune response that protected against subsequent genital challenge with WT Chlamydia muridarum Collectively, our results demonstrate that chlamydia mutants that are profoundly attenuated for specific organ tissues can be derived and demonstrate that live-attenuated vaccine strains that infect the GI tract, but do not elicit genital tract disease, could be used to protect against chlamydia genital tract infection and disease.IMPORTANCE Chlamydia is the most common sexually transmitted bacterial infection in the United States. Most chlamydia genital infections resolve without serious consequences; however, untreated infection in women can cause pelvic inflammatory disease and infertility. Antibiotics are very effective in treating chlamydia, but most genital infections in both men and women are asymptomatic and go undiagnosed. Therefore, there is a critical need for an effective vaccine. In this work, we show that a mutant chlamydia strain, having substantially reduced virulence for genital infection, colonizes the gastrointestinal tract and produces robust immunity to genital challenge with fully virulent wild-type chlamydia. These results are an important advance in understanding chlamydial virulence and provide compelling evidence that safe and effective live-attenuated chlamydia vaccines may be feasible.


Subject(s)
Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Cross Protection/immunology , Gastroenteritis/immunology , Reproductive Tract Infections/immunology , Animals , Antibodies, Bacterial/immunology , Chlamydia Infections/microbiology , Chlamydia muridarum/genetics , Disease Models, Animal , Disease Susceptibility , Female , Gastroenteritis/microbiology , Gastrointestinal Tract/microbiology , Genitalia/microbiology , Genome, Bacterial , Host-Pathogen Interactions/immunology , Mice , Mutation , Polymorphism, Single Nucleotide , Reproductive Tract Infections/microbiology , Virulence
20.
Infect Immun ; 89(1)2020 12 15.
Article in English | MEDLINE | ID: mdl-33077619

ABSTRACT

Gastrointestinal (GI) colonization with group B Streptococcus (GBS) is an important precursor to late-onset (LO) disease in infants. The host-pathogen interactions that mediate progression to invasive disease remain unknown due, in part, to a paucity of robust model systems. Passively acquired maternal GBS-specific antibodies protect newborns from early-onset disease, yet their impact on GI colonization and LO disease is unexplored. Using murine models of both perinatal and postnatal GBS acquisition, we assessed the kinetics of GBS GI colonization, progression to invasive disease, and the role of GBS-specific IgG production in exposed offspring and juvenile mice at age 12 and 14 days, respectively. We defined LO disease as >7 days of life in the perinatal model. We studied the impact of maternal immunization using a whole-cell GBS vaccine on the duration of intestinal colonization and progression to invasive disease after postnatal GBS exposure in offspring. Animals exhibit sustained GI colonization following both perinatal and postnatal exposure to GBS, with 21% and 27%, respectively, developing invasive disease. Intestinal colonization with GBS induces an endogenous IgG response within 20 days of exposure. Maternal vaccination with whole-cell GBS induces production of GBS-specific IgG in dams that is vertically transmitted to their offspring but does not decrease the duration of GBS intestinal colonization or reduce LO mortality following postnatal GBS exposure. Both perinatal and postnatal murine models of GBS acquisition closely recapitulate the human disease state, in which GBS colonizes the intestine and causes LO disease. We demonstrate both endogenous production of anti-GBS IgG in juvenile mice and vertical transfer of antibodies to offspring following maternal vaccination. These models serve as a platform to study critical host-pathogen interactions that mediate LO GBS disease.


Subject(s)
Antibodies, Bacterial/immunology , Gastroenteritis/immunology , Gastroenteritis/microbiology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus agalactiae/immunology , Age Factors , Animals , Antibodies, Bacterial/blood , Disease Models, Animal , Disease Susceptibility , Gastroenteritis/mortality , Gastroenteritis/pathology , Host-Pathogen Interactions/immunology , Immunization , Mice , Streptococcal Infections/mortality , Streptococcal Infections/pathology , Streptococcal Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...