Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 926
Filter
1.
Front Immunol ; 15: 1278197, 2024.
Article in English | MEDLINE | ID: mdl-38803492

ABSTRACT

Background: Primary immunodeficiencies are heritable defects in immune system function. Antibody deficiency is the most common form of primary immunodeficiency in humans, can be caused by abnormalities in both the development and activation of B cells, and may result from B-cell-intrinsic defects or defective responses by other cells relevant to humoral immunity. Inflammatory gastrointestinal complications are commonly observed in antibody-deficient patients, but the underlying immune mechanisms driving this are largely undefined. Methods: In this study, several mouse strains reflecting a spectrum of primary antibody deficiency (IgA-/-, Aicda-/-, CD19-/- and JH -/-) were used to generate a functional small-bowel-specific cellular atlas using a novel high-parameter flow cytometry approach that allows for the enumeration of 59 unique cell subsets. Using this cellular atlas, we generated a direct and quantifiable estimate of immune dysregulation. This estimate was then used to identify specific immune factors most predictive of the severity of inflammatory disease of the small bowel (small bowel enteropathy). Results: Results from our experiments indicate that the severity of primary antibody deficiency positively correlates with the degree of immune dysregulation that can be expected to develop in an individual. In the SI of mice, immune dysregulation is primarily explained by defective homeostatic responses in T cell and invariant natural killer-like T (iNKT) cell subsets. These defects are strongly correlated with abnormalities in the balance between protein (MHCII-mediated) versus lipid (CD1d-mediated) antigen presentation by intestinal epithelial cells (IECs) and intestinal stem cells (ISCs), respectively. Conclusions: Multivariate statistical approaches can be used to obtain quantifiable estimates of immune dysregulation based on high-parameter flow cytometry readouts of immune function. Using one such estimate, we reveal a previously unrecognized tradeoff between iNKT cell activation and type 1 immunity that underlies disease in the small bowel. The balance between protein/lipid antigen presentation by ISCs may play a crucial role in regulating this balance and thereby suppressing inflammatory disease in the small bowel.


Subject(s)
Disease Models, Animal , Flow Cytometry , Intestine, Small , Animals , Mice , Flow Cytometry/methods , Intestine, Small/immunology , Intestine, Small/pathology , Mice, Knockout , Primary Immunodeficiency Diseases/immunology , Primary Immunodeficiency Diseases/genetics , Mice, Inbred C57BL , B-Lymphocytes/immunology , Intestinal Diseases/immunology , Intestinal Diseases/pathology
2.
Int Immunopharmacol ; 135: 112271, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38762923

ABSTRACT

The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signal has drawn much consideration due to its sensitivity to DNA in innate immune mechanisms. Activation of the cGAS-STIN signaling pathway induces the production of interferon and inflammatory cytokines, resulting in immune responses, or inflammatory diseases. The intestinal tract is a vital organ for the body's nutrition absorption, recent studies have had various points of view on the job of cGAS-STING pathway in various intestinal sicknesses. Therefore, understanding its role and mechanism in the intestinal environment can help to develop new strategies for the treatment of intestinal diseases. This article examines the mechanism of the cGAS-STING pathway and its function in inflammatory bowel disease, intestinal cancer, and long-injury ischemia-reperfusion, lists the current medications that target it for the treatment of intestinal diseases, and discusses the impact of intestinal flora on this signaling pathway, to offer a theoretical and scientific foundation for upcoming targeted therapies for intestinal disorders via the cGAS-STING pathway.


Subject(s)
Intestinal Diseases , Membrane Proteins , Nucleotidyltransferases , Signal Transduction , Animals , Humans , Immunity, Innate , Intestinal Diseases/immunology , Intestinal Diseases/metabolism , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism
3.
Nat Rev Gastroenterol Hepatol ; 21(6): 428-443, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38467885

ABSTRACT

The gastrointestinal tract is an immunologically rich organ, containing complex cell networks and dense lymphoid structures that safeguard this large absorptive barrier from pathogens, contribute to tissue physiology and support mucosal healing. Simultaneously, the immune system must remain tolerant to innocuous dietary antigens and trillions of normally beneficial microorganisms colonizing the intestine. Indeed, a dysfunctional immune response in the intestine underlies the pathogenesis of numerous local and systemic diseases, including inflammatory bowel disease, food allergy, chronic enteric infections or cancers. Here, we discuss group 3 innate lymphoid cells (ILC3s), which have emerged as orchestrators of tissue physiology, immunity, inflammation, tolerance and malignancy in the gastrointestinal tract. ILC3s are abundant in the developing and healthy intestine but their numbers or function are altered during chronic disease and cancer. The latest studies provide new insights into the mechanisms by which ILC3s fundamentally shape intestinal homeostasis or disease pathophysiology, and often this functional dichotomy depends on context and complex interactions with other cell types or microorganisms. Finally, we consider how this knowledge could be harnessed to improve current treatments or provoke new opportunities for therapeutic intervention to promote gut health.


Subject(s)
Immunity, Innate , Lymphocytes , Humans , Immunity, Innate/immunology , Lymphocytes/immunology , Intestines/immunology , Intestinal Mucosa/immunology , Animals , Intestinal Diseases/immunology , Inflammatory Bowel Diseases/immunology
4.
Front Immunol ; 13: 822754, 2022.
Article in English | MEDLINE | ID: mdl-35154141

ABSTRACT

Wild pigs usually showed high tolerance and resistance to several diseases in the wild environment, suggesting that the gut bacteria of wild pigs could be a good source for discovering potential probiotic strains. In our study, wild pig feces were sequenced and showed a higher relative abundance of the genus Lactobacillus (43.61% vs. 2.01%) than that in the domestic pig. A total of 11 lactic acid bacteria (LAB) strains including two L. rhamnosus, six L. mucosae, one L. fermentum, one L. delbrueckii, and one Enterococcus faecalis species were isolated. To investigate the synergistic effects of mixed probiotics strains, the mixture of 11 LAB strains from an intestinal ecology system was orally administrated in mice for 3 weeks, then the mice were challenged with Escherichia coli ATCC 25922 (2 × 109 CFU) and euthanized after challenge. Mice administrated with LAB strains showed higher (p < 0.05) LAB counts in feces and ileum. Moreover, alterations of specific bacterial genera occurred, including the higher (p < 0.05) relative abundance of Butyricicoccus and Clostridium IV and the lower (p < 0.05) abundance of Enterorhabdus in mice fed with mixed LAB strains. Mice challenged with Escherichia coli showed vacuolization of the liver, lower GSH in serum, and lower villus to the crypt proportion and Claudin-3 level in the gut. In contrast, administration of mixed LAB strains attenuated inflammation of the liver and gut, especially the lowered IL-6 and IL-1ß levels (p < 0.05) in the gut. Our study highlighted the importance of gut bacterial diversity and the immunomodulation effects of LAB strains mixture from wild pig in gut health.


Subject(s)
Escherichia coli Infections/therapy , Intestinal Diseases/therapy , Lactobacillales/physiology , Probiotics/pharmacology , Animals , Escherichia coli/drug effects , Escherichia coli Infections/immunology , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Immunity/drug effects , Intestinal Diseases/immunology , Intestinal Diseases/metabolism , Intestinal Diseases/microbiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lactobacillales/isolation & purification , Male , Mice , Mice, Inbred C57BL , Probiotics/therapeutic use , Sus scrofa
5.
J Sci Food Agric ; 102(3): 1066-1075, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34309869

ABSTRACT

BACKGROUND: Inflammation-related diseases present a significant public health problem. Ginger is a flavoring spice and medicinal herb with anti-inflammatory activity. This study investigated the preventive effects of ginger extract (GE) and its main bioactive component, 6-gingerol (6G), on lipopolysaccharide (LPS)-induced intestinal barrier dysfunction and liver injury in mice. RESULTS: GE and 6G were orally administered to mice for seven consecutive days before LPS administration. After 24 h, the mice were sacrificed. GE and 6G were found to significantly reverse LPS-induced inflammation in the mouse ileum by modifying the NF-κB pathway. They also alleviated apoptosis in the ileum by downregulating Bax and cytochrome c gene expression and by inhibiting the caspase-3 pathway. Through the aforementioned mechanisms, GE and 6G restored the intestinal barrier by increasing ZO-1 and claudin-1 protein expressions. Gut-derived LPS induced inflammation and apoptosis in the liver; these effects were markedly reversed through GE and 6G treatment. 6G was the most abundant component in GE, as evidenced through liquid chromatography-mass spectrometry, and accounted for >50% of total gingerols and shogaols in GE. CONCLUSION: The current results support the use of GE and 6G as dietary supplements to protect against gut-derived endotoxemia-associated inflammatory response and disorders. © 2021 Society of Chemical Industry.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Catechols/administration & dosage , Fatty Alcohols/administration & dosage , Intestinal Diseases/drug therapy , Liver Diseases/drug therapy , Plant Extracts/administration & dosage , Zingiber officinale/chemistry , Animals , Apoptosis/drug effects , Humans , Intestinal Diseases/immunology , Intestinal Diseases/physiopathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/injuries , Lipopolysaccharides/adverse effects , Liver/drug effects , Liver/immunology , Liver/injuries , Liver Diseases/immunology , Liver Diseases/physiopathology , Male , Mice , Mice, Inbred ICR
6.
Am J Surg Pathol ; 46(1): 89-96, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34081038

ABSTRACT

Approximately 20% of patients with symptomatic syndrome-associated coronavirus-2 (SARS-CoV-2) infection have gastrointestinal bleeding and/or diarrhea. Most are managed without endoscopic evaluation because the risk of practitioner infection outweighs the value of biopsy analysis unless symptoms are life-threatening. As a result, much of what is known about the gastrointestinal manifestations of coronavirus disease-2019 (COVID-19) has been gleaned from surgical and autopsy cases that suffer from extensive ischemic injury and/or poor preservation. There are no detailed reports describing any other gastrointestinal effects of SARS-CoV-2 even though >3,000,000 people have died from COVID-19 worldwide. The purpose of this study is to report the intestinal findings related to SARS-CoV-2 infection by way of a small case series including one with evidence of direct viral cytopathic effect and 2 with secondary injury attributed to viral infection. Infection can be confirmed by immunohistochemical stains directed against SARS-CoV-2 spike protein, in situ hybridization for spike protein-encoding RNA, and ultrastructural visualization of viruses within the epithelium. It induces cytoplasmic blebs and tufted epithelial cells without inflammation and may not cause symptoms. In contrast, SARS-CoV-2 infection can cause gastrointestinal symptoms after the virus is no longer detected, reflecting systemic activation of cytokine and complement cascades rather than direct viral injury. Reversible mucosal ischemia features microvascular injury with hemorrhage, small vessel thrombosis, and platelet-rich thrombi. Systemic cytokine elaboration and dysbiosis likely explain epithelial cell injury that accompanies diarrheal symptoms. These observations are consistent with clinical and in vitro data and contribute to our understanding of the protean manifestations of COVID-19.


Subject(s)
COVID-19/pathology , Intestinal Diseases/pathology , Intestinal Diseases/virology , Intestines/pathology , Intestines/virology , Adult , Aged , Aged, 80 and over , Biomarkers/metabolism , Biopsy , COVID-19/diagnosis , COVID-19/immunology , Cytokines/metabolism , Gastrointestinal Hemorrhage/diagnosis , Gastrointestinal Hemorrhage/immunology , Gastrointestinal Hemorrhage/pathology , Gastrointestinal Hemorrhage/virology , Humans , Intestinal Diseases/diagnosis , Intestinal Diseases/immunology , Intestines/immunology , Ischemia/diagnosis , Ischemia/immunology , Ischemia/pathology , Ischemia/virology , Male , Thrombosis/diagnosis , Thrombosis/immunology , Thrombosis/pathology , Thrombosis/virology
7.
Front Immunol ; 12: 704836, 2021.
Article in English | MEDLINE | ID: mdl-34650552

ABSTRACT

Intestinal ischemia/reperfusion (I/R) injury is a grave condition with high morbidity and mortality. We previously confirmed that intestinal I/R induces intestinal flora disorders and changes in metabolites, but the role of different metabolites in intestinal I/R injury is currently unclear. Based on targeted metabolic sequencing, pravastatin (PA) was determined to be a metabolite of the gut microbiota. Further, intestinal I/R model mice were established through superior mesenteric artery obstruction. In addition, a co-culture model of small intestinal organoids and type II innate lymphoid cells (ILC2s) was subjected to hypoxia/reoxygenation (H/R) to simulate an intestinal I/R model. Moreover, correlation analysis between the PA level in preoperative feces of patients undergoing cardiopulmonary bypass and the indices of postoperative intestinal I/R injury was carried out. IL-33-deficient mice, ILC2-deleted mice, and anti-IL-13 neutralizing antibodies were also used to explore the potential mechanism through which PA attenuates intestinal I/R injury. We demonstrated that PA levels in the preoperative stool of patients undergoing cardiopulmonary bypass were negatively correlated with the indices of postoperative intestinal I/R injury. Furthermore, PA alleviated intestinal I/R injury and improved the survival of mice. We further showed that PA promotes IL-13 release from ILC2s by activating IL-33/ST2 signaling to attenuate intestinal I/R injury. In addition, IL-13 promoted the self-renewal of intestinal stem cells by activating Notch1 and Wnt signals. Overall, results indicated that the gut microbial metabolite PA can attenuate intestinal I/R injury by promoting the release of IL-13 from ILC2s via IL-33/ST2 signaling, revealing a novel mechanism of and therapeutic strategy for intestinal I/R injury.


Subject(s)
Gastrointestinal Microbiome/immunology , Immunity, Innate , Interleukin-1 Receptor-Like 1 Protein/immunology , Interleukin-13/immunology , Interleukin-33/immunology , Intestinal Diseases/immunology , Lymphocytes/immunology , Pravastatin/immunology , Animals , Disease Models, Animal , Humans , Interleukin-1 Receptor-Like 1 Protein/genetics , Interleukin-13/genetics , Interleukin-33/genetics , Intestinal Diseases/genetics , Male , Mice , Mice, Knockout , Reperfusion Injury
8.
Oxid Med Cell Longev ; 2021: 5541222, 2021.
Article in English | MEDLINE | ID: mdl-34712384

ABSTRACT

Many gut disease etiologies are attributed to the presence of robust inflammatory cell recruitment. The recruitment of neutrophils plays a vital role in inflammatory infiltration. Neutrophils have various antimicrobial effector mechanisms, including phagocytosis, oxidative burst, and degranulation. It is suggested that neutrophils could release neutrophil extracellular traps (NETs) to kill pathogens. However, recent evidence indicates that neutrophil infiltration within the gut is associated with disrupted local immunological microenvironment and impaired epithelial barrier. Growing evidence implies that NETs are involved in the progression of many diseases, including cancer, diabetes, thrombosis, and autoimmune disease. Increased NET formation was found in acute or chronic conditions, including infection, sterile inflammation, cancer, and ischemia/reperfusion injury (IRI). Here, we present a comprehensive review of recent advances in the understanding of NETs, focusing on their effects in gut disease. We also discuss NETs as a potential therapeutic target in gut disease.


Subject(s)
Extracellular Traps/metabolism , Intestinal Diseases/metabolism , Intestines/metabolism , Neutrophil Infiltration , Neutrophils/metabolism , Animals , Extracellular Traps/drug effects , Gastrointestinal Agents/therapeutic use , Humans , Intestinal Diseases/drug therapy , Intestinal Diseases/immunology , Intestinal Diseases/pathology , Intestines/drug effects , Intestines/immunology , Intestines/pathology , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Neutrophils/immunology
9.
J Immunol Res ; 2021: 7183136, 2021.
Article in English | MEDLINE | ID: mdl-34485536

ABSTRACT

The intestinal mucosal immune barrier protects the host from the invasion of foreign pathogenic microorganisms. Immune cells and cytokines in the intestinal mucosa maintain local and systemic homeostasis by participating in natural and adaptive immunity. Deficiency of the intestinal mucosal immune barrier is associated with a variety of intestinal illnesses. Exosomes are phospholipid bilayer nanovesicles that allow cell-cell communication by secreting physiologically active substances including proteins, lipids, transcription factors, mRNAs, micro-RNAs (miRNAs), and long noncoding RNAs (lncRNAs). Exosomal lncRNAs are involved in immune cell differentiation and the modulation of the immune response. This review briefly introduces the potential role of exosomal lncRNAs in the intestinal mucosal immune barrier and discusses their relevance to intestinal illnesses.


Subject(s)
Exosomes/metabolism , Immunity, Mucosal/genetics , Intestinal Diseases/immunology , Intestinal Mucosa/immunology , RNA, Long Noncoding/metabolism , Animals , Disease Models, Animal , Gene Expression Regulation/immunology , Humans , Intestinal Diseases/diagnosis , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Severity of Illness Index
10.
Mucosal Immunol ; 14(6): 1381-1392, 2021 11.
Article in English | MEDLINE | ID: mdl-34420043

ABSTRACT

The SARS-CoV-2 pandemic has so far claimed over three and a half million lives worldwide. Though the SARS-CoV-2 mediated disease COVID-19 has first been characterized by an infection of the upper airways and the lung, recent evidence suggests a complex disease including gastrointestinal symptoms. Even if a direct viral tropism of intestinal cells has recently been demonstrated, it remains unclear, whether gastrointestinal symptoms are caused by direct infection of the gastrointestinal tract by SARS-CoV-2 or whether they are a consequence of a systemic immune activation and subsequent modulation of the mucosal immune system. To better understand the cause of intestinal symptoms we analyzed biopsies of the small intestine from SARS-CoV-2 infected individuals. Applying qRT-PCR and immunohistochemistry, we detected SARS-CoV-2 RNA and nucleocapsid protein in duodenal mucosa. In addition, applying imaging mass cytometry and immunohistochemistry, we identified histomorphological changes of the epithelium, which were characterized by an accumulation of activated intraepithelial CD8+ T cells as well as epithelial apoptosis and subsequent regenerative proliferation in the small intestine of COVID-19 patients. In summary, our findings indicate that intraepithelial CD8+ T cells are activated upon infection of intestinal epithelial cells with SARS-CoV-2, providing one possible explanation for gastrointestinal symptoms associated with COVID-19.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , Duodenum/immunology , Immunity, Mucosal , Intestinal Diseases/immunology , Intestinal Mucosa/immunology , Intraepithelial Lymphocytes/immunology , Lymphocyte Activation , SARS-CoV-2/immunology , Adult , Aged , Animals , Apoptosis , CD8-Positive T-Lymphocytes/virology , COVID-19/pathology , COVID-19/virology , Case-Control Studies , Cell Proliferation , Chlorocebus aethiops , Duodenum/pathology , Duodenum/virology , Female , Host-Pathogen Interactions , Humans , Intestinal Diseases/pathology , Intestinal Diseases/virology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Intraepithelial Lymphocytes/virology , Male , Re-Epithelialization , SARS-CoV-2/pathogenicity , Vero Cells , Viral Load
11.
Mucosal Immunol ; 14(6): 1335-1346, 2021 11.
Article in English | MEDLINE | ID: mdl-34326478

ABSTRACT

Intestinal inflammation can be accompanied by osteoporosis, but their relationship, mediated by immune responses, remains unclear. Here, we investigated a non-IgE-mediated food-allergic enteropathy model of ovalbumin (OVA) 23-3 mice expressing OVA-specific T-cell-receptor transgenes. Mesenteric lymph nodes (MLNs) and their pathogenic CD4+T cells were important to enteropathy occurrence and exacerbation when the mice were fed an egg-white (EW) diet. EW-fed OVA23-3 mice also developed bone loss and increased CD44hiCD62LloCD4+T cells in the MLNs and bone marrow (BM); these changes were attenuated by MLN, but not spleen, resection. We fed an EW diet to F1 cross offspring from OVA23-3 mice and a mouse line expressing the photoconvertible protein KikGR to track MLN CD4+T cells. Photoconverted MLN CD44hiCD62LloCD4+T cells migrated predominantly to the BM; pit formation assay proved their ability to promote bone damage via osteoclasts. Significantly greater expression of IL-4 mRNA in MLN CD44hiCD62LloCD4+T cells and bone was observed in EW-fed OVA23-3 mice. Anti-IL-4 monoclonal antibody injection canceled bone loss in the primary inflammation phase in EW-fed mice, but less so in the chronic phase. This novel report shows the specific inflammatory relationship, via Th2-dominant-OVA-specific T cells and IL-4 production, between MLNs and bone, a distant organ, in food-allergic enteropathy.


Subject(s)
Bone Resorption/etiology , CD4-Positive T-Lymphocytes/physiology , Food Hypersensitivity/complications , Food Hypersensitivity/immunology , Interleukin-4/genetics , Intestinal Diseases/immunology , Lymph Nodes/immunology , Memory T Cells/physiology , Animals , Biomarkers , Bone Resorption/diagnostic imaging , Bone Resorption/metabolism , Bone Resorption/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Food Hypersensitivity/metabolism , Immunophenotyping , Interleukin-4/metabolism , Intestinal Diseases/complications , Intestinal Diseases/metabolism , Lymph Nodes/metabolism , Mesentery , Mice , Models, Biological
12.
J Fish Dis ; 44(10): 1619-1637, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34237181

ABSTRACT

Anti-nutritional factors in dietary components can have a negative impact on the intestinal barrier. Here, we present soya bean-induced changes in the intestine of juvenile zebrafish and the effect of yeast ß-glucan through a transcriptomic approach. The inclusion of soya bean meal affected the expression of several intestinal barrier function-related genes like arl4ca, rab25b, rhoub, muc5ac, muc5d, clcn2c and cltb in zebrafish. Several metabolic genes like cyp2x10.2, cyp2aa2, aldh3a2b, crata, elovl4, elovl6, slc51a, gpat2 and ATP-dependent peptidase activity (lonrf, clpxb) were altered in the intestinal tissue. The expression of immune-related genes like nlrc3, nlrp12, gimap8, prdm1 and tph1a, and genes related to cell cycle, DNA damage and DNA repair (e.g. spo11, rad21l1, nabp1b, spata22, tdrd9) were also affected in the soya bean fed group. Furthermore, our study suggests the plausible effect of yeast ß-glucan through the modulation of several genes that regulate immune responses and barrier integrity. Our findings indicate a subdued inflammation in juvenile zebrafish fed soya bean meal and the efficacy of ß-glucan to counter these subtle inflammatory responses.


Subject(s)
Fish Diseases/prevention & control , Glycine max/chemistry , Inflammation/veterinary , Intestinal Diseases/prevention & control , Polysaccharides/metabolism , Transcriptome/drug effects , Zebrafish , Animal Feed/analysis , Animals , Diet/veterinary , Dietary Supplements/analysis , Fish Diseases/immunology , Gene Expression Regulation/drug effects , Inflammation/immunology , Inflammation/prevention & control , Intestinal Diseases/immunology , Intestines , Polysaccharides/administration & dosage , Polysaccharides/chemistry , Saccharomyces cerevisiae/chemistry
13.
Food Funct ; 12(14): 6271-6282, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34105571

ABSTRACT

A polysaccharide from Cordyceps sinensis (NCSP) was reported to attenuate intestinal injury and regulate the balance of T helper (Th)1/Th2 cells in immunosuppressed mice. However, whether it influences Th17 and regulatory T (Treg) cells as well as gut ecology remains unknown. In the present study, the intestinal injury mouse model was also established by intraperitoneal injection of cyclophosphamide (Cy) for three consecutive days. NCSP was found to increase the number of CD4+ T cells, stimulate the secretion of interleukins (IL)-17 and IL-21, and the expression of transcription factor (retinoic acid-related orphan receptor (ROR)-γt). The levels of transforming growth factor (TGF)-ß3 and transcription factor (forkhead box (Fox)p-3) were increased in NCSP-treated groups. Moreover, NCSP upregulated the mRNA expression of toll like receptors (TLR-2, -6 and -9), while it downregulated the TLR-4 expression. In addition, NCSP modulated the intestinal microbiota composition and increased the levels of SCFAs. These findings indicated that NCSP may enhance intestinal immunity and have the potential to become a prebiotic to regulate intestinal microbiota.


Subject(s)
Cordyceps/chemistry , Fungal Polysaccharides/pharmacology , Gastrointestinal Microbiome/drug effects , Intestinal Diseases/drug therapy , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cyclophosphamide/adverse effects , Female , Interleukin-17/immunology , Interleukins/immunology , Intestinal Diseases/immunology , Mice , Mice, Inbred BALB C , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Regulatory/drug effects , Th1 Cells/immunology , Th17 Cells/immunology , Transcription Factors/metabolism
14.
Vet Immunol Immunopathol ; 237: 110271, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34044267

ABSTRACT

Canine chronic enteropathy (CE) is a group of immunogenetic disorders of unclear etiology characterized by chronic or recurrent gastrointestinal signs and inflammation. Diagnosis of CE subtypes by treatment response is a lengthy and challenging process, particularly in refractory cases of the disease. Given known association of dog leukocyte antigen (DLA) class II genotype and various immunogenetic disorders between and across breeds, this study was designed to examine the potential of determining susceptibility to refractory CE through identification of risk and protective genotypes in French bulldogs and miniature dachshunds-two popular dog breeds in Japan. Sequence-based genotyping of three DLA class II genes in 29 French bulldogs and 30 miniature dachshunds with refractory CE revealed a protective haplotype DLA-DRB1*002:01-DQA1*009:01-DQB1*001:01 against CE in French bulldogs (OR 0.09, 95 % CI 0.01-0.71, p = 0.0084). No statistical difference was noted between miniature dachshund cases and controls. These findings, largely disparate from a previous study on German shepherd dogs in the UK, were taken as possible indication of etiological differences in the refractory CE noted between and within breeds, and by extension, the potential of identifying such disease heterogeneity by DLA typing. The DLA-DQA1/DQB1 haplotype, protective against CE in our French bulldogs, has been reported as protective in various immune-mediated disorders such as Doberman hepatitis (Dyggve et al., 2011). Likewise, the DLA-DRB1*006:01 risk allele for Doberman hepatitis was noted in more French bulldogs with CE compared to controls, in line with reports on genotypes associated with both risk and protection being shared across various autoimmune diseases and breeds. These findings support an immunogenetic basis to the French bulldog-CE in our analysis, calling for further DLA studies working with larger samples and different breeds towards phenotypic clarification that may aid in early diagnosis, treatment, and prophylaxis through epigenetic approaches and breeding.


Subject(s)
Histocompatibility Antigens Class II/genetics , Intestinal Diseases/veterinary , Alleles , Animals , Chronic Disease , Dog Diseases/immunology , Dogs , Female , Genetic Association Studies , Genotype , Intestinal Diseases/genetics , Intestinal Diseases/immunology , Male , Species Specificity
15.
Front Immunol ; 12: 644982, 2021.
Article in English | MEDLINE | ID: mdl-33815399

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative therapy for many hematological disorders and autoimmune diseases, but acute graft-versus-host disease (aGVHD) has remained a major obstacle that limits allo-HSCT and exhibits a daunting mortality rate. The gastrointestinal system is among the most common sites affected by aGVHD. Experimental advances in the field of intestinal microbiota research enhanced our understanding - not only of the quantity and diversity of intestinal microbiota - but also their association with homeostasis of the immune system and disease pathogenesis, including that of aGVHD. Meanwhile, ever-growing clinical evidence suggest that the intestinal microbiota is dysregulated in patients who develop aGVHD and that the imbalance may affect clinical outcomes, indicating a potential predictive role for microbiota dysregulation in aGVHD severity and prognosis. The current animal and human studies investigating the intestinal microbiota in aGVHD and the understanding of the influence and management of the microbiota in the clinic are reviewed herein. Taken together, monitoring and remodeling the intestinal microecology following allo-HSCT may provide us with promising avenues for diagnosing, preventing or treating aGVHD in the clinic.


Subject(s)
Gastrointestinal Microbiome/immunology , Graft vs Host Disease , Hematologic Diseases , Hematopoietic Stem Cell Transplantation , Intestinal Diseases , Intestines , Animals , Graft vs Host Disease/immunology , Graft vs Host Disease/microbiology , Graft vs Host Disease/mortality , Hematologic Diseases/immunology , Hematologic Diseases/microbiology , Hematologic Diseases/mortality , Hematologic Diseases/therapy , Humans , Intestinal Diseases/immunology , Intestinal Diseases/microbiology , Intestinal Diseases/mortality , Intestines/immunology , Intestines/microbiology , Transplantation, Homologous
16.
J Exp Med ; 218(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33760921

ABSTRACT

The diverse enteric viral communities that infect microbes and the animal host collectively constitute the gut virome. Although recent advances in sequencing and analysis of metaviromes have revealed the complexity of the virome and facilitated discovery of new viruses, our understanding of the enteric virome is still incomplete. Recent studies have uncovered how virome-host interactions can contribute to beneficial or detrimental outcomes for the host. Understanding the complex interactions between enteric viruses and the intestinal immune system is a prerequisite for elucidating their role in intestinal diseases. In this review, we provide an overview of the enteric virome composition and summarize recent findings about how enteric viruses are sensed by and, in turn, modulate host immune responses during homeostasis and disease.


Subject(s)
Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Homeostasis/immunology , Immune System/immunology , Virome/immunology , Animals , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/virology , Host Microbial Interactions/immunology , Humans , Intestinal Diseases/immunology , Intestinal Diseases/virology , Virome/physiology , Virus Diseases/immunology , Virus Diseases/virology
17.
AAPS J ; 23(2): 32, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33629139

ABSTRACT

As one of the most important metabolites of vitamin A, all-trans retinoic acid (RA) plays a crucial role in regulating immune responses. RA has been shown to promote the differentiation of naïve T and B cells and perform diverse functions in the presence of different cytokines. RA also induces gut tropic lymphocytes through upregulating the expression of chemokine (C-C motif) receptor 9 (CCR9) and α4ß7 integrin. In addition, RA promotes the expression of the enzyme retinal dehydrogenase (RALDH) on dendritic cells, which in turn strengthens the ability to synthesize RA. Due to the insolubility of RA, proper formulation design can maximize its ability to improve immune responses for vaccines. Recent studies have developed some formulations co-loading RA and antigen, which can effectively imprint lymphocytes gut homing properties and induce intestine immune responses as well as systemic responses through parenteral administration, providing a promising direction for the protection against mucosal infections. Here, we review the mechanism and effects of RA on lymphocyte differentiation and gut homing, and recent progress of RA delivery systems to improve mucosal immune responses.


Subject(s)
Drug Carriers/chemistry , Immunity, Mucosal/drug effects , Intestinal Diseases/prevention & control , Tretinoin/administration & dosage , Vaccines/administration & dosage , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Drug Compounding/methods , Humans , Immunogenicity, Vaccine , Intestinal Diseases/immunology , Intestinal Diseases/microbiology , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Mice , Solubility , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tretinoin/chemistry , Tretinoin/immunology , Vaccine Excipients/chemistry , Vaccines/chemistry , Vaccines/immunology
18.
J Autoimmun ; 118: 102609, 2021 03.
Article in English | MEDLINE | ID: mdl-33607573

ABSTRACT

Immune-mediated enteropathies are caused by excessive reactions of the intestinal immune system towards non-pathogenic molecules. Enteropathy leads to malabsorption-related symptoms and include (severe) chronic diarrhea, weight loss and vitamin deficiencies. Parenteral feeding and immunosuppressive therapy are needed in severe cases. Celiac disease has long been recognized as the most common immune-mediated enteropathy in adults, but the spectrum of immune-mediated enteropathies has been expanding. Histological and clinical features are sometimes shared among these enteropathies, and therefore it may be challenging to differentiate between them. Here, we provide an overview of immune-mediated enteropathies focused on clinical presentation, establishing diagnosis, immunopathogenesis, and treatment options.


Subject(s)
Immune System Diseases/immunology , Immunosuppression Therapy/methods , Intestinal Diseases/immunology , Parenteral Nutrition , Animals , Clinical Trials as Topic , Disease Models, Animal , Humans , Immune System Diseases/diagnosis , Immune System Diseases/pathology , Immune System Diseases/therapy , Immunity, Mucosal , Intestinal Diseases/diagnosis , Intestinal Diseases/pathology , Intestinal Diseases/therapy , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Treatment Outcome
19.
Int J Mol Sci ; 22(3)2021 Jan 30.
Article in English | MEDLINE | ID: mdl-33573273

ABSTRACT

After their synthesis from cholesterol in hepatic tissues, bile acids (BAs) are secreted into the intestinal lumen. Most BAs are subsequently re-absorbed in the terminal ileum and are transported back for recycling to the liver. Some of them, however, reach the colon and change their physicochemical properties upon modification by gut bacteria, and vice versa, BAs also shape the composition and function of the intestinal microbiota. This mutual interplay of both BAs and gut microbiota regulates many physiological processes, including the lipid, carbohydrate and energy metabolism of the host. Emerging evidence also implies an important role of this enterohepatic BA circuit in shaping mucosal colonization resistance as well as local and distant immune responses, tissue physiology and carcinogenesis. Subsequently, disrupted interactions of gut bacteria and BAs are associated with many disorders as diverse as Clostridioides difficile or Salmonella Typhimurium infection, inflammatory bowel disease, type 1 diabetes, asthma, metabolic syndrome, obesity, Parkinson's disease, schizophrenia and epilepsy. As we cannot address all of these interesting underlying pathophysiologic mechanisms here, we summarize the current knowledge about the physiologic and pathogenic interplay of local site microbiota and the enterohepatic BA metabolism using a few selected examples of liver and gut diseases.


Subject(s)
Bile Acids and Salts/metabolism , Gastrointestinal Microbiome/physiology , Intestinal Diseases/metabolism , Liver Diseases/metabolism , Animals , Carbohydrate Metabolism/physiology , Disease Models, Animal , Energy Metabolism/physiology , Host Microbial Interactions/physiology , Humans , Intestinal Diseases/immunology , Intestinal Diseases/microbiology , Intestinal Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Lipid Metabolism/physiology , Liver/immunology , Liver/metabolism , Liver/pathology , Liver Diseases/immunology , Liver Diseases/microbiology , Liver Diseases/pathology
20.
Vet Immunol Immunopathol ; 234: 110216, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33636544

ABSTRACT

The pathogenesis of chronic inflammatory enteropathies (CIE) in dogs involves dysregulated innate immune responses. The receptor for advanced glycation end products (RAGE), a pattern recognition receptor, plays a role in chronic inflammation. Abrogation of proinflammatory RAGE signaling by ligand binding (e.g., S100/calgranulins) to soluble RAGE (sRAGE) might also be a novel therapeutic avenue. Serum sRAGE levels are decreased in canine CIE, but gastrointestinal tissue RAGE expression has not been investigated in dogs. Thus, the study aimed to evaluate the gastrointestinal mucosal RAGE expression in dogs with CIE. Further, the potential binding of RAGE to canine S100/calgranulin ligands was investigated. Epithelial RAGE expression was quantified in gastrointestinal (gastric, duodenal, ileal, and colonic) biopsies from 12 dogs with CIE and 9 healthy control dogs using confocal laser scanning microscopy. RAGE expression was compared between both groups of dogs and was tested for an association with patient characteristics, clinical variables, histologic lesion severity, and biomarkers of extra-gastrointestinal disease, systemic or gastrointestinal inflammation, function, or protein loss. Statistical significance was set at p < 0.05. RAGE:S100/calgranulin binding was assessed by immunoassay and electrophoretic techniques. RAGE expression was detected in all 59 biopsies from diseased and healthy control dogs evaluated. Epithelial RAGE expression in the duodenum and colon was significantly higher in dogs with CIE than in healthy controls (p < 0.04). Compared to healthy controls, RAGE expression in dogs with CIE also tended to be higher in the ileum but lower in the stomach. A slight (statistically not significant) shift towards more basal intestinal epithelial RAGE expression was detected in CIE dogs. Serum sRAGE was proportional to epithelial RAGE expression in the duodenum (p < 0.04), and RAGE expression in the colon inversely correlated with biomarkers of protein loss in serum (both p < 0.04). Several histologic morphologic and inflammatory lesion criteria and markers of inflammation (serum C-reactive protein and fecal calprotectin concentration) were related to epithelial RAGE expression in the duodenum, ileum, and/or colon. in vitro canine RAGE:S100A12 binding appeared more pronounced than RAGE:S100A8/A9 binding. This study showed a dysregulation of epithelial RAGE expression along the gastrointestinal tract in dogs with CIE. Compensatory regulations in the sRAGE/RAGE axis are an alternative explanation for these findings. The results suggest that RAGE signaling plays a role in dogs with CIE, but higher anti-inflammatory decoy receptor sRAGE levels paralleled RAGE overexpression. Canine S100/calgranulins were demonstrated to be ligands for RAGE.


Subject(s)
Biopsy/veterinary , Dog Diseases/genetics , Gene Expression , Inflammation/veterinary , Intestinal Diseases/genetics , Intestinal Diseases/immunology , Receptor for Advanced Glycation End Products/genetics , Animals , Dog Diseases/immunology , Dogs , Female , Gastrointestinal Tract/pathology , Humans , Inflammation/genetics , Male , Receptor for Advanced Glycation End Products/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...