Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 276
Filter
1.
Int J Mol Sci ; 24(17)2023 Aug 22.
Article in English | MEDLINE | ID: mdl-37685869

ABSTRACT

ITGAM-ITGAX (rs11150612, rs11574637), VAV3 rs17019602, CARD9 rs4077515, DEFA (rs2738048, rs10086568), and HORMAD2 rs2412971 are mucosal immune defence polymorphisms, that have an impact on IgA production, described as risk loci for IgA nephropathy (IgAN). Since IgAN and Immunoglobulin-A vasculitis (IgAV) share molecular mechanisms, with the aberrant deposit of IgA1 being the main pathophysiologic feature of both entities, we assessed the potential influence of the seven abovementioned polymorphisms on IgAV pathogenesis. These seven variants were genotyped in 381 Caucasian IgAV patients and 997 matched healthy controls. No statistically significant differences were observed in the genotype and allele frequencies of these seven polymorphisms when the whole cohort of IgAV patients and those with nephritis were compared to controls. Similar genotype and allele frequencies of all polymorphisms were disclosed when IgAV patients were stratified according to the age at disease onset or the presence/absence of gastrointestinal or renal manifestations. Likewise, no ITGAM-ITGAX and DEFA haplotype differences were observed when the whole cohort of IgAV patients, along with those with nephritis and controls, as well as IgAV patients, stratified according to the abovementioned clinical characteristics, were compared. Our results suggest that mucosal immune defence polymorphisms do not represent novel genetic risk factors for IgAV pathogenesis.


Subject(s)
Glomerulonephritis, IGA , IgA Vasculitis , Immunity, Mucosal , Nephritis , Humans , CD11c Antigen , Gene Frequency , Genotype , Glomerulonephritis, IGA/genetics , IgA Vasculitis/genetics , Polymorphism, Genetic , Immunity, Mucosal/genetics
2.
Biochem Genet ; 60(6): 2299-2312, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35334059

ABSTRACT

Viruses are the most common and abundant organisms in the marine environment. To better understand how cetaceans have adapted to this virus-rich environment, we compared cetacean virus-responsive genes to those from terrestrial mammals. We identified virus-responsive gene sequences in seven species of cetaceans, which we compared with orthologous sequences in seven terrestrial mammals. As a result of evolution analysis using the branch model and the branch-site model, 21 genes were selected using at least one model. IFN-ε, an antiviral cytokine expressed at mucous membranes, and its receptor IFNAR1 contain cetacean-specific amino acid substitutions that might change the interaction between the two proteins and lead to regulation of the immune system against viruses. Cetacean-specific amino acid substitutions in IL-6, IL-27, and the signal transducer and activator of transcription (STAT)1 are also predicted to alter the mucosal immune response of cetaceans. Since mucosal membranes are the first line of defense against the external environment and are involved in immune tolerance, our analysis of cetacean virus-responsive genes suggests that genes with cetacean-specific mutations in mucosal immunity-related genes play an important role in the protection and/or regulation of immune responses against viruses.


Subject(s)
Cetacea , Immunity, Mucosal , Animals , Immunity, Mucosal/genetics , Phylogeny , Cetacea/genetics , Mammals , Adaptation, Physiological
3.
Front Immunol ; 12: 769775, 2021.
Article in English | MEDLINE | ID: mdl-34804060

ABSTRACT

The crosstalk between the immune system and microbiota drives an amazingly complex mutualistic symbiosis. In mammals, the upper respiratory tract acts as a gateway for pathogen invasion, and the dynamic interaction between microbiota and mucosal immunity on its surface can effectively prevent disease development. However, the relationship between virus-mediated mucosal immune responses and microbes in lower vertebrates remains uncharacterized. In this study, we successfully constructed an infection model by intraperitoneally injecting common carp (Cyprinus carpio) with spring viremia of carp virus (SVCV). In addition to the detection of the SVCV in the nose and pharynx of common carp, we also identified obvious histopathological changes following viral infection. Moreover, numerous immune-related genes were significantly upregulated in the nose and pharynx at the peak of SVCV infection, after which the expression levels decreased to levels similar to those of the control group. Transcriptome sequencing results revealed that pathways associated with bacterial infection in the Toll-like receptor pathway and the Nod-like receptor pathway were activated in addition to the virus-related Rig-I-like receptor pathway after SVCV infection, suggesting that viral infection may be followed by opportunistic bacterial infection in these mucosal tissues. Using 16S rRNA gene sequencing, we further identified an upward trend in pathogenic bacteria on the mucosal surface of the nose and pharynx 4 days after SVCV infection, after which these tissues eventually reached new homeostasis. Taken together, our results suggest that the dynamic interaction between mucosal immunity and microbiota promotes the host to a new ecological state.


Subject(s)
Bacteria/immunology , Carps/immunology , Fish Diseases/immunology , Immunity, Mucosal/immunology , Pharynx/immunology , Rhabdoviridae/immunology , Animal Structures/immunology , Animal Structures/microbiology , Animal Structures/virology , Animals , Bacteria/classification , Bacteria/genetics , Carps/microbiology , Carps/virology , Fish Diseases/microbiology , Fish Diseases/virology , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/methods , Homeostasis/genetics , Homeostasis/immunology , Immunity, Mucosal/genetics , Pharynx/microbiology , Pharynx/virology , Phylogeny , RNA, Ribosomal, 16S/genetics , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Rhabdoviridae/genetics , Rhabdoviridae/physiology , Signal Transduction/genetics , Signal Transduction/immunology
4.
PLoS One ; 16(9): e0256657, 2021.
Article in English | MEDLINE | ID: mdl-34492052

ABSTRACT

INTRODUCTION: The etiology of diverticulosis is still poorly understood. However, in patients with diverticulitis, markers of mucosal inflammation and microbiota alterations have been found. The aim of this study was to evaluate potential differences of the gut microbiota composition and mucosal immunity between patients with asymptomatic diverticulosis and controls. METHODS: We performed a prospective study on patients who underwent routine colonoscopy for causes not related to diverticular disease or inflammatory bowel disease. Participants were grouped based on the presence or absence of diverticula. Mucosal biopsies were obtained from the sigmoid and transverse colon. Microbiota composition was analyzed with IS-pro, a 16S-23S based bacterial profiling technique. To predict if patients belonged to the asymptomatic diverticulosis or control group a partial least squares discriminant analysis (PLS-DA) regression model was used. Inflammation was assessed by neutrophil and lymphocyte counts within the taken biopsies. RESULTS: Forty-three patients were enrolled. Intestinal microbiota profiles were highly similar within individuals for all phyla. Between individuals, microbiota profiles differed substantially but regardless of the presence (n = 19) of absence (n = 24) of diverticula. Microbiota diversity in both sigmoid and transverse colon was similar in all participants. We were not able to differentiate between diverticulosis patients and controls with a PLS-DA model. Mucosal lymphocyte counts were comparable among both groups; no neutrophils were detected in any of the studied biopsies. CONCLUSIONS: Microbiota composition and inflammatory markers were comparable among asymptomatic diverticulosis patients and controls. This suggests that the gut microbiota and mucosal inflammation do not play a major role in the pathogenesis of diverticula formation.


Subject(s)
Asymptomatic Diseases/epidemiology , Diverticulum/immunology , Diverticulum/microbiology , Inflammation/microbiology , Aged , Colon, Sigmoid/microbiology , Colon, Sigmoid/pathology , Colonoscopy , Diverticulum/epidemiology , Diverticulum/genetics , Female , Gastrointestinal Microbiome/genetics , Humans , Immunity, Mucosal/genetics , Immunity, Mucosal/immunology , Inflammation/epidemiology , Inflammation/pathology , Male , Middle Aged , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/immunology
5.
Front Immunol ; 12: 730346, 2021.
Article in English | MEDLINE | ID: mdl-34566991

ABSTRACT

Mast cell activators are a novel class of mucosal vaccine adjuvants. The polymeric compound, Compound 48/80 (C48/80), and cationic peptide, Mastoparan 7 (M7) are mast cell activators that provide adjuvant activity when administered by the nasal route. However, small molecule mast cell activators may be a more cost-efficient adjuvant alternative that is easily synthesized with high purity compared to M7 or C48/80. To identify novel mast cell activating compounds that could be evaluated for mucosal vaccine adjuvant activity, we employed high-throughput screening to assess over 55,000 small molecules for mast cell degranulation activity. Fifteen mast cell activating compounds were down-selected to five compounds based on in vitro immune activation activities including cytokine production and cellular cytotoxicity, synthesis feasibility, and selection for functional diversity. These small molecule mast cell activators were evaluated for in vivo adjuvant activity and induction of protective immunity against West Nile Virus infection in BALB/c mice when combined with West Nile Virus envelope domain III (EDIII) protein in a nasal vaccine. We found that three of the five mast cell activators, ST101036, ST048871, and R529877, evoked high levels of EDIII-specific antibody and conferred comparable levels of protection against WNV challenge. The level of protection provided by these small molecule mast cell activators was comparable to the protection evoked by M7 (67%) but markedly higher than the levels seen with mice immunized with EDIII alone (no adjuvant 33%). Thus, novel small molecule mast cell activators identified by high throughput screening are as efficacious as previously described mast cell activators when used as nasal vaccine adjuvants and represent next-generation mast cell activators for evaluation in mucosal vaccine studies.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cell Degranulation/drug effects , Immunity, Mucosal/drug effects , Mast Cells/drug effects , West Nile Fever/prevention & control , West Nile Virus Vaccines/administration & dosage , West Nile virus/pathogenicity , Administration, Intranasal , Animals , Cell Line , Disease Models, Animal , Drug Discovery , Female , High-Throughput Screening Assays , Host-Pathogen Interactions , Immunity, Mucosal/genetics , Immunization , Immunogenicity, Vaccine , Mast Cells/immunology , Mast Cells/virology , Mice, Inbred BALB C , Proof of Concept Study , West Nile Fever/genetics , West Nile Fever/immunology , West Nile Fever/virology , West Nile virus/immunology
6.
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
7.
Int J Mol Sci ; 22(17)2021 Aug 28.
Article in English | MEDLINE | ID: mdl-34502262

ABSTRACT

Intestinal microfold cells (M cells) are a dynamic lineage of epithelial cells that initiate mucosal immunity in the intestine. They are responsible for the uptake and transcytosis of microorganisms, pathogens, and other antigens in the gastrointestinal tract. A mature M cell expresses a receptor Gp2 which binds to pathogens and aids in the uptake. Due to the rarity of these cells in the intestine, their development and differentiation remain yet to be fully understood. We recently demonstrated that polycomb repressive complex 2 (PRC2) is an epigenetic regulator of M cell development, and 12 novel transcription factors including Atoh8 were revealed to be regulated by the PRC2. Here, we show that Atoh8 acts as a regulator of M cell differentiation; the absence of Atoh8 led to a significant increase in the number of Gp2+ mature M cells and other M cell-associated markers such as Spi-B and Sox8. In vitro organoid analysis of RankL treated organoid showed an increase of mature marker GP2 expression and other M cell-associated markers. Atoh8 null mice showed an increase in transcytosis capacity of luminal antigens. An increase in M cell population has been previously reported to be detrimental to mucosal immunity because some pathogens like orally acquired prions have been able to exploit the transcytosis capacity of M cells to infect the host; mice with an increased population of M cells are also susceptible to Salmonella infections. Our study here demonstrates that PRC2 regulated Atoh8 is one of the factors that regulate the population density of intestinal M cell in the Peyer's patch.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Animals , B-Lymphocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/immunology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Immunity, Mucosal/genetics , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Mice , Mice, Knockout , Peyer's Patches/drug effects , Peyer's Patches/metabolism , Primary Cell Culture , RANK Ligand/pharmacology , Receptor Activator of Nuclear Factor-kappa B/pharmacology , T-Lymphocytes/metabolism , Transcytosis/genetics
9.
Dev Comp Immunol ; 124: 104198, 2021 11.
Article in English | MEDLINE | ID: mdl-34237381

ABSTRACT

Since the identification of a functional Cδ gene in ostriches, immunoglobulin (Ig) D has been considered to be an extremely evolutionarily conserved Ig isotype besides the IgM found in all classes of jawed vertebrates. However, in contrast to IgM (which remains stable over evolutionary time), IgD shows considerable structural plasticity among vertebrate species and, moreover, its functions are far from elucidated even in humans and mice. Recently, several studies have shown that high expression of the IgD-B-cell receptor (IgD-BCR) may help physiologically autoreactive B cells survive in peripheral lymphoid tissues thanks to unresponsiveness to self-antigens and help their entry into germinal centers to "redeem" autoreactivity via somatic hypermutation. Other studies have demonstrated that secreted IgD may enhance mucosal homeostasis and immunity by linking B cells with basophils to optimize T-helper-2 cell-mediated responses and to constrain IgE-mediated basophil degranulation. Herein, we review the new discoveries on IgD-encoding genes in jawed vertebrates in the past decade. We also highlight advances in the functions of the IgD-BCR and secreted IgD in humans and mice.


Subject(s)
Genes, Immunoglobulin , Immunoglobulin D/genetics , Animals , B-Lymphocytes/immunology , Gene Expression Regulation/immunology , Humans , Immune Tolerance/genetics , Immunity, Mucosal/genetics , Immunoglobulin D/immunology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Vertebrates/classification , Vertebrates/genetics , Vertebrates/immunology
10.
Commun Biol ; 4(1): 913, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34312491

ABSTRACT

Tissue-resident γδ intraepithelial lymphocytes (IELs) orchestrate innate and adaptive immune responses to maintain intestinal epithelial barrier integrity. Epithelia-specific butyrophilin-like (Btnl) molecules induce perinatal development of distinct Vγ TCR+ IELs, however, the mechanisms that control γδ IEL maintenance within discrete intestinal segments are unclear. Here, we show that Btnl2 suppressed homeostatic proliferation of γδ IELs preferentially in the ileum. High throughput transcriptomic characterization of site-specific Btnl2-KO γδ IELs reveals that Btnl2 regulated the antimicrobial response module of ileal γδ IELs. Btnl2 deficiency shapes the TCR specificities and TCRγ/δ repertoire diversity of ileal γδ IELs. During DSS-induced colitis, Btnl2-KO mice exhibit increased inflammation and delayed mucosal repair in the colon. Collectively, these data suggest that Btnl2 fine-tunes γδ IEL frequencies and TCR specificities in response to site-specific homeostatic and inflammatory cues. Hence, Btnl-mediated targeting of γδ IEL development and maintenance may help dissect their immunological functions in intestinal diseases with segment-specific manifestations.


Subject(s)
Butyrophilins/genetics , Ileum/immunology , Immunity, Innate/genetics , Immunity, Mucosal/genetics , Intraepithelial Lymphocytes/metabolism , Animals , Butyrophilins/metabolism , Female , Mice , Mice, Inbred C57BL
11.
Mucosal Immunol ; 14(5): 1113-1126, 2021 09.
Article in English | MEDLINE | ID: mdl-34158595

ABSTRACT

Despite the impact of childhood diarrhea on morbidity and mortality, our understanding of its sequelae has been significantly hampered by the lack of studies that examine samples across the entire intestinal tract. Infant rhesus macaques are naturally susceptible to human enteric pathogens and recapitulate the hallmarks of diarrheal disease such as intestinal inflammation and growth faltering. Here, we examined intestinal biopsies, lamina propria leukocytes, luminal contents, and fecal samples from healthy infants and those experiencing growth faltering with distant acute or chronic active diarrhea. We show that growth faltering in the presence or absence of active diarrhea is associated with a heightened systemic and mucosal pro-inflammatory state centered in the colon. Moreover, polyclonal stimulation of colonic lamina propria leukocytes resulted in a dampened cytokine response, indicative of immune exhaustion. We also detected a functional and taxonomic shift in the luminal microbiome across multiple gut sites including the migration of Streptococcus and Prevotella species between the small and large intestine, suggesting a decompartmentalization of gut microbial communities. Our studies provide valuable insight into the outcomes of diarrheal diseases and growth faltering not attainable in humans and lays the groundwork to test interventions in a controlled and reproducible setting.


Subject(s)
Diarrhea/metabolism , Dysbiosis/complications , Gastrointestinal Microbiome/immunology , Growth Disorders/etiology , Intestinal Mucosa/immunology , Animals , Biodiversity , Biomarkers , Biopsy , Chronic Disease , Diarrhea/complications , Diarrhea/etiology , Diarrhea/pathology , Disease Models, Animal , Disease Susceptibility , Dysbiosis/immunology , Growth Disorders/metabolism , Immunity, Mucosal/genetics , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lymphocyte Count , Macaca mulatta , Metagenome , Metagenomics/methods , Transcriptome
12.
Dev Comp Immunol ; 124: 104164, 2021 11.
Article in English | MEDLINE | ID: mdl-34129850

ABSTRACT

MicroRNAs could not only regulate posttranscriptional silencing of target genes in eukaryotic organisms, but also have positive effect on their target genes as well. These microRNAs have been reported to be involved in mucosal immune responses to pathogen infection in teleost. Therefore, we constructed the immune-related miRNA-mRNA networks in turbot intestine following Vibrio anguillarum infection. In our results, 1550 differentially expressed (DE) genes and 167 DE miRNAs were identified. 113 DE miRNAs targeting 89 DE mRNAs related to immune response were used to construct miRNA-mRNA interaction networks. Functional analysis showed that target genes were associated with synthesis and degradation of ketone bodies, mucin type O-Glycan biosynthesis, homologous recombination, biotin metabolism, and intestinal immune network for IgA production that were equivalent to the function of IgT and IgM in fish intestine. Finally, 10 DE miRNAs and 7 DE mRNAs were selected for validating the accuracy of high-throughput sequencing results by qRT-PCR. The results of this study will provide valuable information for the elucidation of the regulation mechanisms of miRNA-mRNA interactions involved in disease resistance in teleost mucosal immune system.


Subject(s)
Fish Diseases/genetics , Flatfishes/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , Vibrio Infections/veterinary , Vibrio/physiology , Animals , Disease Resistance/genetics , Disease Resistance/immunology , Fish Diseases/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Flatfishes/immunology , Gene Expression Regulation/immunology , Gene Regulatory Networks/immunology , Immunity, Mucosal/genetics , MicroRNAs/immunology , RNA, Messenger/immunology , Vibrio Infections/genetics , Vibrio Infections/immunology
13.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33822777

ABSTRACT

BACKGROUNDCoronavirus disease 2019 (COVID-19) is more benign in children compared with adults for unknown reasons. This contrasts with other respiratory viruses where disease manifestations are often more severe in children. We hypothesize that a more robust early innate immune response to SARS coronavirus 2 (SARS-CoV-2) protects against severe disease.METHODSClinical outcomes, SARS-CoV-2 viral copies, and cellular gene expression were compared in nasopharyngeal swabs obtained at the time of presentation to the emergency department from 12 children and 27 adults using bulk RNA sequencing and quantitative reverse-transcription PCR. Total protein, cytokines, and anti-SARS-CoV-2 IgG and IgA were quantified in nasal fluid.RESULTSSARS-CoV-2 copies, angiotensin-converting enzyme 2, and TMPRSS2 gene expression were similar in children and adults, but children displayed higher expression of genes associated with IFN signaling, NLRP3 inflammasome, and other innate pathways. Higher levels of IFN-α2, IFN-γ, IP-10, IL-8, and IL-1ß protein were detected in nasal fluid in children versus adults. Children also expressed higher levels of genes associated with immune cells, whereas expression of those associated with epithelial cells did not differ in children versus adults. Anti-SARS-CoV-2 IgA and IgG were detected at similar levels in nasal fluid from both groups. None of the children required supplemental oxygen, whereas 7 adults did (P = 0.03); 4 adults died.CONCLUSIONThese findings provide direct evidence of a more vigorous early mucosal immune response in children compared with adults and suggest that this contributes to favorable clinical outcomes.FUNDINGNIH grants R01 AI134367, UL1 TR002556, T32 AI007501, T32GM007288, P30 AI124414; an Albert Einstein College of Medicine Dean's COVID-19 Pilot Research Award; and the Eric J. Heyer, MD, PhD Translational Research Pilot Project Award.


Subject(s)
COVID-19/immunology , Immunity, Mucosal , SARS-CoV-2 , Adult , Aged , Antibodies, Viral/metabolism , COVID-19/genetics , Child , Child, Preschool , Cytokines/metabolism , Female , Humans , Immunity, Innate/genetics , Immunity, Mucosal/genetics , Infant , Male , Middle Aged , Nasal Mucosa/immunology , Pandemics , SARS-CoV-2/immunology , Transcriptome
14.
Front Immunol ; 12: 625297, 2021.
Article in English | MEDLINE | ID: mdl-33746962

ABSTRACT

Essential oils (EOs) are promising alternatives to chemotherapeutics in animal production due to their immunostimulant, antimicrobial, and antioxidant properties, without associated environmental or hazardous side effects. In the present study, the modulation of the transcriptional immune response (microarray analysis) and microbiota [16S Ribosomal RNA (rRNA) sequencing] in the intestine of the euryhaline fish gilthead seabream (Sparus aurata) fed a dietary supplementation of garlic, carvacrol, and thymol EOs was evaluated. The transcriptomic functional analysis showed the regulation of genes related to processes of proteolysis and inflammatory modulation, immunity, transport and secretion, response to cyclic compounds, symbiosis, and RNA metabolism in fish fed the EOs-supplemented diet. Particularly, the activation of leukocytes, such as acidophilic granulocytes, was suggested to be the primary actors of the innate immune response promoted by the tested functional feed additive in the gut. Fish growth performance and gut microbiota alpha diversity indices were not affected, while dietary EOs promoted alterations in bacterial abundances in terms of phylum, class, and genus. Subtle, but significant alterations in microbiota composition, such as the decrease in Bacteroidia and Clostridia classes, were suggested to participate in the modulation of the intestine transcriptional immune profile observed in fish fed the EOs diet. Moreover, regarding microbiota functionality, increased bacterial sequences associated with glutathione and lipid metabolisms, among others, detected in fish fed the EOs supported the metabolic alterations suggested to potentially affect the observed immune-related transcriptional response. The overall results indicated that the tested dietary EOs may promote intestinal local immunity through the impact of the EOs on the host-microbial co-metabolism and consequent regulation of significant biological processes, evidencing the crosstalk between gut and microbiota in the inflammatory regulation upon administration of immunostimulant feed additives.


Subject(s)
Bacteria/drug effects , Dietary Supplements , Gastrointestinal Microbiome/drug effects , Immunity, Innate/drug effects , Immunity, Mucosal/drug effects , Intestines/drug effects , Oils, Volatile/administration & dosage , Sea Bream , Transcriptome/drug effects , Allyl Compounds/administration & dosage , Animal Feed , Animals , Bacteria/genetics , Bacteria/growth & development , Cymenes/administration & dosage , Diet , Drug Combinations , Gene Expression Profiling , Gene Regulatory Networks/drug effects , Immunity, Innate/genetics , Immunity, Mucosal/genetics , Intestines/immunology , Intestines/microbiology , Oligonucleotide Array Sequence Analysis , Ribotyping , Sea Bream/genetics , Sea Bream/immunology , Sea Bream/metabolism , Sea Bream/microbiology , Sulfides/administration & dosage , Thymol/administration & dosage
15.
Mol Immunol ; 133: 14-22, 2021 05.
Article in English | MEDLINE | ID: mdl-33610122

ABSTRACT

The polymeric immunoglobulin receptor (pIgR) is an important molecule in the mucosal immunity of teleosts. Previous studies have shown that pIgR can bind and transport polymeric immunoglobulins (pIgs), but few studies have focused on the binding of teleost pIgR to bacteria. In this study, we identified a gene encoding pIgR in largemouth bass (Micropterus salmoides). The pIgR gene contained two Ig-like domains (ILDs), which were homologous to ILD1 and ILD5 of mammalian pIgR. Our results showed that largemouth bass pIgR-ILD could combine with IgM. Moreover, we also found that largemouth bass pIgR-ILD could bind to Aeromonas hydrophila and Micrococcus luteus. Further analysis showed that largemouth bass pIgR-ILD could also combine with lipopolysaccharide (LPS), peptidoglycan (PGN) and various saccharides, and reduced binding to bacteria was observed with LPS and PGN treatment, indicating that largemouth bass pIgR could bind to bacteria to prevent infection and that saccharide binding is an important interaction mechanism between pIgR and bacteria. These results collectively demonstrated that largemouth bass pIgR not only combines with IgM but also binds to bacteria by various saccharides.


Subject(s)
Aeromonas hydrophila/immunology , Bass/immunology , Immunoglobulin M/immunology , Micrococcus luteus/immunology , Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/immunology , Amino Acid Sequence , Animals , Base Sequence , Bass/genetics , Fish Diseases/immunology , Immunity, Mucosal/genetics , Immunity, Mucosal/immunology , Lipopolysaccharides/immunology , Peptidoglycan/immunology , Phylogeny , Protein Domains/genetics , Sequence Alignment , Sequence Analysis, DNA
16.
Science ; 371(6526)2021 01 15.
Article in English | MEDLINE | ID: mdl-33446526

ABSTRACT

Human monogenic disorders have revealed the critical contribution of type 17 responses in mucosal fungal surveillance. We unexpectedly found that in certain settings, enhanced type 1 immunity rather than defective type 17 responses can promote mucosal fungal infection susceptibility. Notably, in mice and humans with AIRE deficiency, an autoimmune disease characterized by selective susceptibility to mucosal but not systemic fungal infection, mucosal type 17 responses are intact while type 1 responses are exacerbated. These responses promote aberrant interferon-γ (IFN-γ)- and signal transducer and activator of transcription 1 (STAT1)-dependent epithelial barrier defects as well as mucosal fungal infection susceptibility. Concordantly, genetic and pharmacologic inhibition of IFN-γ or Janus kinase (JAK)-STAT signaling ameliorates mucosal fungal disease. Thus, we identify aberrant T cell-dependent, type 1 mucosal inflammation as a critical tissue-specific pathogenic mechanism that promotes mucosal fungal infection susceptibility in mice and humans.


Subject(s)
Candida albicans/immunology , Candidiasis, Chronic Mucocutaneous/genetics , Candidiasis, Chronic Mucocutaneous/immunology , Immunity, Mucosal/immunology , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Adolescent , Adult , Aged , Animals , Disease Models, Animal , Female , Humans , Immunity, Mucosal/genetics , Immunologic Surveillance/genetics , Immunologic Surveillance/immunology , Interferon-gamma/genetics , Interleukins/genetics , Janus Kinases/genetics , Male , Mice , Mice, Inbred BALB C , Middle Aged , Mouth Mucosa/immunology , Mouth Mucosa/pathology , Receptors, Interleukin-17/genetics , STAT1 Transcription Factor/genetics , T-Lymphocytes/immunology , Young Adult , Interleukin-22
17.
Cell Mol Life Sci ; 78(4): 1191-1206, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32979054

ABSTRACT

Most cervical cancer (CxCa) are related to persistent infection with high-risk human papillomavirus (HR-HPV) in the cervical mucosa, suggesting that an induction of mucosal cell-mediated immunity against HR-HPV oncoproteins can be a promising strategy to fight HPV-associated CxCa. From this perspective, many pre-clinical and clinical trials have proved the potential of lactic acid bacteria (LAB) genetically modified to deliver recombinant antigens to induce mucosal, humoral and cellular immunity in the host. Altogether, the outcomes of these studies suggest that there are several key factors to consider that may offer guidance on improvement protein yield and improving immune response. Overall, these findings showed that oral LAB-based mucosal HPV vaccines expressing inducible surface-anchored antigens display a higher potential to induce particularly specific systemic and mucosal cytotoxic cellular immune responses. In this review, we describe all LAB-based HPV vaccine investigations by reviewing databases from international studies between 2000 and 2020. Our aim is to promote the therapeutic HPV vaccines knowledge and to complete the gaps in this field to empower scientists worldwide to make proper decisions regarding the best strategies for the development of therapeutic HPV vaccines.


Subject(s)
Gastrointestinal Microbiome/genetics , Lactobacillales/genetics , Microorganisms, Genetically-Modified/genetics , Papillomavirus Infections/genetics , Female , Gastrointestinal Microbiome/immunology , Humans , Immunity, Mucosal/genetics , Immunity, Mucosal/immunology , Lactobacillales/immunology , Microorganisms, Genetically-Modified/immunology , Papillomaviridae/drug effects , Papillomaviridae/immunology , Papillomaviridae/pathogenicity , Papillomavirus Infections/immunology , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Papillomavirus Vaccines/immunology , Papillomavirus Vaccines/therapeutic use , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Vagina/immunology , Vagina/microbiology , Vagina/virology
18.
Vet Microbiol ; 251: 108896, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33091794

ABSTRACT

Seasonal influenza is an acute viral infection caused by influenza virus, which is often prevalent in the summer and winter. The influenza virus can infect pigs and poultry. Some literature reports that the influenza virus has an outbreak in summer. The summer climate is characterized by a high humidity and high temperature environment, which is the same as many animal feeding and growing environments. We established a flu animal model under a high temperature and humidity environment during the day to observe the impact of high humidity and high temperature environment on the mice after contracting the influenza virus. Our results indicate that the intestinal flora of 16 s rDNA cultured in High humidity and high temperature environment changes, the intestinal mucosal permeability increases, the expression of pIgR, sIgA, and IgA in the intestinal mucosal immune system decreases, and the NLR immune recognition signaling pathway NOD1 is activated. The expression of related genes such as NOD2, NF-κB, and pIgR increases, which leads to the increase of related inflammatory factors in the vicinity of the intestines, aggravating local inflammation. High humidity and high temperature environment can cause the expression of inflammatory cytokines in the body to rise, causing Th17/Treg immune imbalance, inhibiting Treg maturation and differentiation, and increasing the expression of IL-2, IL-6, and other cytokines, while the expression of IFN-γ and IL-17A decreases. This condition worsens after infection with the influenza virus. Overall, our study found that High humidity and high temperature environment affect the intestinal flora and the body's immune status, thereby aggravating the status of influenza virus infection.


Subject(s)
Environment , Gastrointestinal Microbiome/genetics , Hot Temperature/adverse effects , Humidity/adverse effects , Orthomyxoviridae Infections/immunology , Signal Transduction , Animals , Disease Models, Animal , Female , Immunity, Mucosal/genetics , Immunity, Mucosal/immunology , Lung/virology , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/genetics , Nod1 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Seasons , Swine
19.
J Fish Biol ; 97(5): 1542-1553, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32885862

ABSTRACT

Crucian carp (Carassius auratus) is one of the major freshwater species and important food fish in China. Fish skin acts as the first line of defence against pathogens, yet its molecular and immune mechanism remains unclear. In this study, a de novo transcriptome assembly of C. auratus skin was performed with the Illumina Hiseq 2000 platform. A total of 49,154,776 clean reads were assembled, among which 60,824 (46.86%), 37,103 (28.59%), 43,269 (33.33%) unigenes were annotated against National Center for Biotechnology Information, Gene Onotology and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, respectively. KEGG Orthology categories were significantly involved in immune system (20.50%), signal transduction (18.04%) and mucosal mucin genes (e.g., muc2, muc5AC, muc5B, muc17, muc18). The high expression of muc18 gene was observed in brain; that of muc2 in intestine; and that of muc5AC in skin, liver, spleen, intestine and muscle. Moreover, the potential 28,928 simple sequence repeats with the three most abundant dinucleotide repeat motifs (AC/GT, AG/CT, AT/AT) were detected in C. auratus. To authors' knowledge, this is the first report to describe the transcriptome analysis of C. auratus skin, and the outcome of this study contributed to the understanding of mucosal immune response of the skin and molecular markers in cyprinid species.


Subject(s)
Goldfish/genetics , Goldfish/metabolism , Mucins/genetics , Mucins/metabolism , Skin/metabolism , Transcriptome , Animals , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Immunity, Mucosal/genetics , Microsatellite Repeats/genetics , Tissue Distribution
20.
J Fish Dis ; 43(12): 1541-1552, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32924190

ABSTRACT

Cryptocaryon irritans, a pathogen model for fish mucosal immunity, causes skin mucosal and systematic humoral immune response. Where and how MHC II antigen presentation occurs in fish infected with C. irritans remain unknown. In this study, the full-length cDNA of the grouper cysteine protease CTSS was cloned. The expression distributions of six genes (CTSB, CTSL, CTSS, GILT, MHC IIA and MHC IIB) involved in MHC II antigen presentation pathway were tested. These genes were highly expressed in systematic immune tissues and skin and gill mucosal-associated immune tissues. All six genes were upregulated in skin at most time points. Five genes expected CTSS was upregulated in spleen at most time points. CTSB, CTSL and MHC IIA were upregulated in the gill and head kidney at some time points. These results indicate that the presentation of MHC II antigen intensively occurred in local infected skin and gill. Spleen, not head kidney, had the most extensive systematic antigen presentation. In skin, six genes most likely peaked at day 2, earlier than in spleen (5-7 days), marking an earlier skin antibody peak than any recorded in serum previously. This significant and earlier mucosal antigen presentation indicates that specific immune response occurs in local mucosal tissues.


Subject(s)
Bass , Ciliophora Infections/immunology , Fish Diseases/parasitology , Major Histocompatibility Complex/genetics , Animals , Antigens, Protozoan , Fish Diseases/genetics , Fish Diseases/immunology , Fish Proteins/genetics , Gene Expression Regulation/immunology , Hymenostomatida/physiology , Immunity, Humoral , Immunity, Mucosal/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...