Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Cytokine ; 151: 155794, 2022 03.
Article in English | MEDLINE | ID: mdl-35030468

ABSTRACT

In a prospective cohort study of 77 children with severe pneumonia from two hospitals in Uganda, we assessed soluble T cell immunoglobulin and mucin-domain containing protein 3 (sTIM-3) levels at hospital admission and their association with pneumonia severity and subsequent mortality. sTIM-3 levels were positively correlated with the Respiratory Index of Severity in Children (RISC) (ρ = 0.35, p = 0.0017), sTIM-3 levels were higher in children who required transfer to a tertiary hospital (p = 0.014) and in fatal cases (p = 0.011). In summary, sTIM-3 is associated with disease severity and predictive of mortality in childhood pneumonia in resource-limited settings.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Pneumonia , Child , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Immunoglobulins/metabolism , Mucin-3/metabolism , Pneumonia/metabolism , Prospective Studies , Severity of Illness Index , T-Lymphocytes/metabolism
2.
Int J Mol Sci ; 23(20)2022 Oct 12.
Article in English | MEDLINE | ID: mdl-36293034

ABSTRACT

Malignant pleural effusion (MPE) provides a liquid tumor microenvironment model that includes cancer cells and immune cells. However, the characteristics of tumor antigen-specific CD8+ T cells have not been investigated in detail. Here, we analyzed MPE samples taken from a patient with pancreatic cancer who received a dendritic cell vaccine targeting Wilms' Tumor 1 (WT1) antigen over the disease course (two points at MPE1st and 2nd, two months after MPE1st). Epithelial cell adhesion molecule (EpCAM)+ cancer cells (PD-L1- or T cell immunoglobulin mucin-3, TIM-3-), both PD-1 or TIM-3 positive CD8+ T cells, and CD14+CD68+CD163+TIM-3+ macrophages increased from the MPE1st to MPE2nd. The ratio of WT1-specific cytotoxic lymphocytes (WT1-CTLs) to MPE CD8+ T cells and IFN-γ secretion of WT1-CTLs were reduced with disease progression. Coincidentally, the fraction of central memory T (TCM) of WT1-CTLs was decreased. On the other hand, CD8+ T cells in response to SMAD4P130L, which is homogeneously expressed in EpCAM+ cancer cells, were detected using in vitro expansion with the HLA-A*11:01 restrictive SVCVNLYH neoantigen. Furthermore, the CD8+ T cell response to SMAD4P130L was diminished following remarkably decreased numbers of CD8+ TCM in MPE samples. In conclusion, CD8+ T cells responding to WT1 or SMAD4P130L neoantigen expressed in EpCAM+ pancreatic cancer cells were detected in MPE. A tumor antigen-specific immune response would provide novel insight into the MPE microenvironment.


Subject(s)
Pancreatic Neoplasms , Pleural Effusion, Malignant , Vaccines , Humans , Epithelial Cell Adhesion Molecule/metabolism , CD8-Positive T-Lymphocytes , B7-H1 Antigen/metabolism , Hepatitis A Virus Cellular Receptor 2/metabolism , WT1 Proteins , Programmed Cell Death 1 Receptor/metabolism , Mucin-3/metabolism , Pancreatic Neoplasms/pathology , Immunoglobulins/metabolism , Vaccines/metabolism , HLA-A Antigens , Tumor Microenvironment , Smad4 Protein/metabolism
3.
Microbiol Immunol ; 64(4): 280-295, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31907968

ABSTRACT

Mucin is a glycoprotein that is the primary component of the mucus overlaying the epithelial tissues. Because mucin functions as a first line of the innate immune system, Pseudomonas aeruginosa appears to require interaction with mucin to establish infection in the host. However, the interactions between P. aeruginosa and mucin have been poorly understood. In this study, using in vivo expression technology (IVET), we attempted to identify mucin-inducible promoters that are likely to be involved in the establishment of P. aeruginosa infection. The IVET analysis revealed that the genes encoding glycosidases, sulfatases, and peptidases that are thought to be required for the utilization of mucin as a nutrient are present in 13 genes downstream of the identified promoters. Our results indicated that, among them, sdsA1 encoding a secreted sulfatase plays a central role in the degradation of mucin. It was then demonstrated that disruption of sdsA1 leads to a decreased release of sulfate from mucin and sulfated sugars. Furthermore, the sdsA1 mutant showed a reduction in the ability of mucin gel penetration and an attenuation of virulence in leukopenic mice compared with the wild-type strain. Collectively, these results suggest that SdsA1 plays an important role as a virulence factor of P. aeruginosa.


Subject(s)
Bacterial Proteins/immunology , Mucin-3/metabolism , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/enzymology , Sulfatases/immunology , Virulence Factors/immunology , Animals , Male , Mice , Mice, Inbred ICR , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Swine , Virulence
4.
J Immunol ; 197(1): 233-43, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27259857

ABSTRACT

Viruses are frequent causes of lower respiratory infection (LRI). Programmed cell death-1 (PD-1) signaling contributes to pulmonary CD8(+) T cell (TCD8) functional impairment during acute viral LRI, but the role of TCD8 impairment in viral clearance and immunopathology is unclear. We now find that human metapneumovirus infection induces virus-specific lung TCD8 that fail to produce effector cytokines or degranulate late postinfection, with minimally increased function even in the absence of PD-1 signaling. Impaired lung TCD8 upregulated multiple inhibitory receptors, including PD-1, lymphocyte activation gene 3 (LAG-3), T cell Ig mucin 3, and 2B4. Moreover, coexpression of these receptors continued to increase even after viral clearance, with most virus-specific lung TCD8 expressing three or more inhibitory receptors on day 14 postinfection. Viral infection also increased expression of inhibitory ligands by both airway epithelial cells and APCs, further establishing an inhibitory environment. In vitro Ab blockade revealed that multiple inhibitory receptors contribute to TCD8 impairment induced by either human metapneumovirus or influenza virus infection. In vivo blockade of T cell Ig mucin 3 signaling failed to enhance TCD8 function or reduce viral titers. However, blockade of LAG-3 in PD-1-deficient mice restored TCD8 effector functions but increased lung pathology, indicating that LAG-3 mediates lung TCD8 impairment in vivo and contributes to protection from immunopathology during viral clearance. These results demonstrate that an orchestrated network of pathways modifies lung TCD8 functionality during viral LRI, with PD-1 and LAG-3 serving prominent roles. Lung TCD8 impairment may prevent immunopathology but also contributes to recurrent lung infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype/immunology , Lung/immunology , Metapneumovirus/immunology , Orthomyxoviridae Infections/immunology , Paramyxoviridae Infections/immunology , Respiratory Tract Infections/immunology , Animals , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/virology , Cells, Cultured , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mucin-3/metabolism , Programmed Cell Death 1 Receptor/genetics , Respiratory Tract Infections/virology , Signal Transduction , Signaling Lymphocytic Activation Molecule Family/metabolism , Lymphocyte Activation Gene 3 Protein
5.
Int J Mol Sci ; 17(11)2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27834807

ABSTRACT

The mucus layer covering the gastrointestinal (GI) epithelium is critical in selecting and maintaining homeostatic interactions with our gut bacteria. However, the molecular details of these interactions are not well understood. Here, we provide mechanistic insights into the adhesion properties of the canonical mucus-binding protein (MUB), a large multi-repeat cell-surface adhesin found in Lactobacillus inhabiting the GI tract. We used atomic force microscopy to unravel the mechanism driving MUB-mediated adhesion to mucins. Using single-molecule force spectroscopy we showed that MUB displayed remarkable adhesive properties favouring a nanospring-like adhesion model between MUB and mucin mediated by unfolding of the multiple repeats constituting the adhesin. We obtained direct evidence for MUB self-interaction; MUB-MUB followed a similar binding pattern, confirming that MUB modular structure mediated such mechanism. This was in marked contrast with the mucin adhesion behaviour presented by Galectin-3 (Gal-3), a mammalian lectin characterised by a single carbohydrate binding domain (CRD). The binding mechanisms reported here perfectly match the particular structural organization of MUB, which maximizes interactions with the mucin glycan receptors through its long and linear multi-repeat structure, potentiating the retention of bacteria within the outer mucus layer.


Subject(s)
Adhesins, Bacterial/chemistry , Galectin 3/chemistry , Limosilactobacillus reuteri/metabolism , Mucin-3/chemistry , Recombinant Proteins/chemistry , Adhesins, Bacterial/isolation & purification , Adhesins, Bacterial/metabolism , Animals , Bacterial Adhesion , Culture Media, Conditioned/chemistry , Galectin 3/genetics , Galectin 3/metabolism , Gene Expression , Humans , Intestinal Mucosa/chemistry , Limosilactobacillus reuteri/growth & development , Microscopy, Atomic Force , Models, Molecular , Mucin-3/isolation & purification , Mucin-3/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Swine
6.
Am J Physiol Cell Physiol ; 307(2): C162-8, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24848113

ABSTRACT

N-acyl-homoserine lactones (AHL) are quorum-sensing molecules in bacteria that play important roles in regulating virulence gene expression in pathogens such as Pseudomonas aeruginosa. The present study compared responses between undifferentiated and differentiated Caco-2 cells to N-(3-oxododecanoyl)-L-homoserine lactone (3-oxo-C12-HSL). A low concentration of 3-oxo-C12-HSL (30 µM) is sufficient to reduce viability accompanied by apoptosis via the suppression of phosphorylation by Akt in undifferentiated Caco-2 cells. The suppression of Akt phosphorylation appears specific in 3-oxo-C12-HSL, because other AHLs did not influence the phosphorylation status of Akt. The reduced viability induced by 3-oxo-C12-HSL was partially recovered by constitutively active Akt overexpression in undifferentiated Caco-2 cells. Since mucin is considered a vital component of the gut barrier, we investigated whether mucin protects cellular functions induced by 3-oxo-C12-HSL in undifferentiated Caco-2 cells. The results showed that mucin protected undifferentiated Caco-2 cells from apoptosis induced by 3-oxo-C12-HSL. 3-Oxo-C12-HSL did not induce cell death in differentiated Caco-2 cells that expressed higher levels of mucin 3 (MUC3) than undifferentiated Caco-2 cells. In addition, 3-oxo-C12-HSL promoted cell death in undifferentiated Caco-2 cells transfected with MUC3 siRNA and reduced MUC3 expression in undifferentiated Caco-2 cells. Therefore, MUC3 might be responsible for the survival of undifferentiated intestinal epithelial cells in the presence of 3-oxo-C12-HSL through regulating Akt phosphorylation. In conclusion, 3-oxo-C12-HSL might influence the survival of undifferentiated intestinal epithelial cells as well as interactions between these cells and pathogens.


Subject(s)
4-Butyrolactone/analogs & derivatives , Apoptosis/drug effects , Epithelial Cells/metabolism , Homoserine/analogs & derivatives , Intestinal Mucosa/cytology , Mucin-3/metabolism , Proto-Oncogene Proteins c-akt/metabolism , 4-Butyrolactone/pharmacology , Caco-2 Cells , Caspase 3/genetics , Caspase 3/metabolism , Gene Expression Regulation , Homoserine/pharmacology , Humans , Phosphorylation
7.
Infect Immun ; 82(2): 509-21, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24478067

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) is a leading cause of death due to diarrheal illness among young children in developing countries, and there is currently no effective vaccine. Many elements of ETEC pathogenesis are still poorly defined. Here we demonstrate that YghJ, a secreted ETEC antigen identified in immunoproteomic studies using convalescent patient sera, is required for efficient access to small intestinal enterocytes and for the optimal delivery of heat-labile toxin (LT). Furthermore, YghJ is a highly conserved metalloprotease that influences intestinal colonization of ETEC by degrading the major mucins in the small intestine, MUC2 and MUC3. Genes encoding YghJ and its cognate type II secretion system (T2SS), which also secretes LT, are highly conserved in ETEC and exist in other enteric pathogens, including other diarrheagenic E. coli and Vibrio cholerae bacteria, suggesting that this mucin-degrading enzyme may represent a shared virulence feature of these important pathogens.


Subject(s)
Enterotoxigenic Escherichia coli/enzymology , Enterotoxigenic Escherichia coli/metabolism , Epithelial Cells/microbiology , Escherichia coli Proteins/metabolism , Metalloproteases/metabolism , Mucin-2/metabolism , Mucin-3/metabolism , Animals , Cell Line , Disease Models, Animal , Escherichia coli Infections/microbiology , Humans , Hydrolysis , Mice , Virulence Factors/metabolism
8.
J Cell Sci ; 124(Pt 18): 3074-83, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21852426

ABSTRACT

The transmembrane mucins in the enterocyte are type 1 transmembrane proteins with long and rigid mucin domains, rich in proline, threonine and serine residues that carry numerous O-glycans. Three of these mucins, MUC3, MUC12 and MUC17 are unique in harboring C-terminal class I PDZ motifs, making them suitable ligands for PDZ proteins. A screening of 123 different human PDZ domains for binding to MUC3 identified a strong interaction with the PDZ protein GOPC (Golgi-associated PDZ and coiled-coil motif-containing protein). This interaction was mediated by the C-terminal PDZ motif of MUC3, binding to the single GOPC PDZ domain. GOPC is also a binding partner for cystic fibrosis transmembrane conductance regulator (CFTR) that directs CFTR for degradation. Overexpression of GOPC downregulated the total levels of MUC3, an effect that was reversed by introducing CFTR. The results suggest that CFTR and MUC3 compete for binding to GOPC, which in turn can regulate levels of these two proteins. For the first time a direct coupling between mucins and the CFTR channel is demonstrated, a finding that will shed further light on the still poorly understood relationship between cystic fibrosis and the mucus phenotype of this disease.


Subject(s)
Carrier Proteins/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Membrane Proteins/metabolism , Mucin-3/metabolism , Voltage-Dependent Anion Channels/metabolism , Adaptor Proteins, Signal Transducing , Animals , Binding, Competitive/genetics , CHO Cells , Carrier Proteins/genetics , Computational Biology , Cricetinae , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Enterocytes/metabolism , Enterocytes/pathology , Feedback, Physiological , Golgi Matrix Proteins , Humans , Membrane Proteins/genetics , Membrane Transport Proteins , Mucin-3/genetics , PDZ Domains/genetics , Transgenes/genetics , Voltage-Dependent Anion Channels/genetics
9.
Afr Health Sci ; 23(3): 607-615, 2023 Sep.
Article in English | MEDLINE | ID: mdl-38357157

ABSTRACT

Background: To investigate the expression of Th17, T lymphocyte immunoglobulin mucin 3 (TIM-3+) cells and their related cytokines in atrial fibrillation (AF) and their clinical significance. Methodology: A total of 90 patients with AF were divided into paroxysmal group (n=45) and chronic group (n=45), and 45 healthy volunteers were selected as the control group. The proportion of Th17 cells and Tim-3 + cells in the peripheral blood were detected. The concentrations of related cytokines in peripheral blood serum were determined. The correlation between Th17 / Tim-3+ cells and related cytokines was analysed. Results: Compared with the control group, the proportion of Th17 cells and the concentration of related cytokines (IL-17, IL-6 and Matrix metalloproteinase (MMP9)) in peripheral blood of patients with paroxysmal and chronic AF increased significantly, while the proportion of tim3 + cells and the concentration of related cytokines decreased significantly. Compared with the paroxysmal group, the proportion of Th17 cells and the concentration of related cytokines in the peripheral blood of patients in the chronic group increased significantly, while the proportion of tim3 + cells and the concentration of related cytokines decreased significantly. Conclusion: Th17 / Tim-3 + cell balance is involved in AF, and can be used as a target for AF treatment.


Subject(s)
Atrial Fibrillation , Hepatitis A Virus Cellular Receptor 2 , Humans , Hepatitis A Virus Cellular Receptor 2/metabolism , Mucin-3/metabolism , Atrial Fibrillation/etiology , Cytokines/metabolism , Th17 Cells/metabolism , Th17 Cells/pathology , Immunoglobulins/metabolism
10.
J Immunol ; 184(4): 1918-30, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20083673

ABSTRACT

T cell/transmembrane, Ig, and mucin (TIM) proteins, identified using a congenic mouse model of asthma, critically regulate innate and adaptive immunity. TIM-1 and TIM-4 are receptors for phosphatidylserine (PtdSer), exposed on the surfaces of apoptotic cells. Herein, we show with structural and biological studies that TIM-3 is also a receptor for PtdSer that binds in a pocket on the N-terminal IgV domain in coordination with a calcium ion. The TIM-3/PtdSer structure is similar to that of TIM-4/PtdSer, reflecting a conserved PtdSer binding mode by TIM family members. Fibroblastic cells expressing mouse or human TIM-3 bound and phagocytosed apoptotic cells, with the BALB/c allelic variant of mouse TIM-3 showing a higher capacity than the congenic C.D2 Es-Hba-allelic variant. These functional differences were due to structural differences in the BC loop of the IgV domain of the TIM-3 polymorphic variants. In contrast to fibroblastic cells, T or B cells expressing TIM-3 formed conjugates with but failed to engulf apoptotic cells. Together these findings indicate that TIM-3-expressing cells can respond to apoptotic cells, but the consequence of TIM-3 engagement of PtdSer depends on the polymorphic variants of and type of cell expressing TIM-3. These findings establish a new paradigm for TIM proteins as PtdSer receptors and unify the function of the TIM gene family, which has been associated with asthma and autoimmunity and shown to modulate peripheral tolerance.


Subject(s)
Alleles , Apoptosis/immunology , Genetic Variation/immunology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mucin-3/genetics , Phagocytosis/immunology , Phosphatidylserines/metabolism , Amino Acid Sequence , Animals , Apoptosis/genetics , Cell Line , Cell Line, Tumor , Hepatitis A Virus Cellular Receptor 2 , Humans , Membrane Proteins/physiology , Mice , Mice, Congenic , Mice, Inbred BALB C , Molecular Sequence Data , Mucin-3/metabolism , Mucin-3/physiology , Multigene Family/immunology , NIH 3T3 Cells , Phagocytosis/genetics
11.
Pathol Int ; 62(6): 381-90, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22612506

ABSTRACT

The differentiation of intraductal papilloma (IDP) in the breast from ductal carcinoma in situ (DCIS) is sometimes difficult. Fifty papillary lesions (25 DCIS and 25 IDP) were immunohistochemically examined using a panel of antibodies, including CK5/6, ER, p63, Ki-67, chromogranin A, synaptophysin, neuron specific enolase, CD56, MUC1, MUC3, CD44, p21, p27, and p53. The immunohistochemical staining pattern of each antibody was evaluated using the Allred scoring system. Then, the area under curve (AUC) for each antibody was computed by receiver operating characteristic (ROC) analysis. DCIS typically showed high scores for ER and MUC3 reactivity compared with IDP, and the AUC for ER and MUC3 were 0.941 and 0.908, respectively. In contrast, IDP showed high scores for CK5/6 and p63 reactivity compared with DCIS, and the AUC for CK5/6 and p63 were 1.00 and 0.954, respectively. We devised a 'Differential Index' (DI) using the following formula: [S(ER) + S(MUC3)]/[S(CK5/6) + S(p63) + 1]. The distributions of the DI for IDP and DCIS did not overlap when the cutoff value was placed arbitrarily at DI = 1.0. From these results, it is concluded that a panel of four CK5/6, ER, p63, and MUC3 antibodies provide valuable information for differentiating IDP from DCIS.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Intraductal, Noninfiltrating/diagnosis , Neoplasm Proteins/metabolism , Papilloma, Intraductal/diagnosis , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Breast Neoplasms/surgery , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/surgery , Diagnosis, Differential , Female , Humans , Immunoenzyme Techniques , Keratin-5/metabolism , Keratin-6/metabolism , Mucin-3/metabolism , Papilloma, Intraductal/metabolism , Papilloma, Intraductal/surgery , Predictive Value of Tests , ROC Curve , Receptors, Estrogen/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
12.
Am J Physiol Gastrointest Liver Physiol ; 301(4): G656-66, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21737776

ABSTRACT

Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in premature infants. NEC is believed to occur when intestinal bacteria invade the intestinal epithelial layer, causing subsequent inflammation and tissue necrosis. Mucins are produced and secreted by epithelial goblet cells as a key component of the innate immune system and barrier function of the intestinal tract that help protect against bacterial invasion. To better understand the role of mucins in NEC, we quantified the number of mucus-containing small intestinal goblet cells present in infants with NEC and found they had significantly fewer goblet cells and Paneth cells compared with controls. To test whether inflammation has a developmentally dependent effect on intestinal goblet cells, TNF-α was injected into mice at various stages of intestinal development. TNF-α caused a loss of mucus-containing goblet cells only in immature mice and induced Muc2 and Muc3 mRNA upregulation only in mature ileum. Only minimal changes were seen in apoptosis and in expression of markers of goblet cell differentiation. TNF-α increased small intestinal mucus secretion and goblet cell hypersensitivity to prostaglandin E2 (PGE(2)), a known mucus secretagogue produced by macrophages. These TNF-α-induced changes in mucus mRNA levels required TNF receptor 2 (TNFR2), whereas TNF-α-induced loss of mucus-positive goblet cells required TNFR1. Our findings of developmentally dependent TNF-α-induced alterations on intestinal mucus may help explain why NEC is predominantly found in premature infants, and TNF-α-induced alterations of the intestinal innate immune system and barrier functions may play a role in the pathogenesis of NEC itself.


Subject(s)
Enterocolitis, Necrotizing/pathology , Intestine, Small/pathology , Receptors, Tumor Necrosis Factor, Type I/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Goblet Cells/metabolism , Goblet Cells/pathology , Humans , Infant, Newborn , Infant, Premature , Intestinal Mucosa/pathology , Intestine, Small/drug effects , Mice , Mucin-2/metabolism , Mucin-3/metabolism , Mucins , Mucus/metabolism , Paneth Cells/metabolism , Paneth Cells/pathology , Proto-Oncogene Proteins c-ets/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/physiology
13.
Pediatr Res ; 69(3): 206-11, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21135754

ABSTRACT

Upon ingestion, probiotics may act to protect the host through a number of protective mechanisms including modulation of genes involved in intestinal innate mucosal defense such as epithelial cell-derived mucin glycoproteins and inhibitor of apoptosis proteins. To determine the specificity of effect and sustainability of response in vivo, Lactobacillus plantarum 299v (Lp299v), Lactobacillus rhamnosus R0011 (LrR0011), and Bifidobacterium bifidum R0071 (BbR0071) were added repeatedly or intermittently to the drinking water of Sprague-Dawley rats. After killing the rats via CO2 suffocation, Muc2, Muc3, neuronal apoptosis inhibitor protein (NAIP), human inhibitor of apoptosis protein 1/cellular inhibitor of apoptosis 2 (HIAP1/cIAP2), and human inhibitor of apoptosis protein 2/cellular inhibitor of apoptosis 1 (HIAP2/cIAP1) mRNA and protein levels were analyzed via RT-PCR and immunohistochemistry. Live Lp299v, BbR0071, and LrR0011 increased Muc3 protein and mRNA expression in jejunum and ileum. Heat-killed and a nonadherent derivative of Lp299v failed to induce Muc3 expression. Lp299v did induce expression of HIAP2/cIAP1 and NAIP expression. Muc3 mucin expression was elevated for 5 d after oral administration of Lp299v; however, this effect was not sustained despite ongoing daily ingestion of a probiotic. Intermittent pulse ingestion of probiotics, however, was found to repeatedly increase Muc3 expression. We conclude that selected probiotics can induce protective genes of mucosal intestinal epithelial cells, an effect that is reproducible with pulse probiotic administration.


Subject(s)
Intestine, Small/drug effects , Intestine, Small/metabolism , Mucin-3/metabolism , Probiotics/pharmacology , Animals , Bifidobacterium/metabolism , Gene Expression , Humans , Lactobacillus plantarum/metabolism , Lacticaseibacillus rhamnosus/metabolism , Male , Mucin-2/genetics , Mucin-2/metabolism , Mucin-3/genetics , Probiotics/administration & dosage , Rats , Rats, Sprague-Dawley , Up-Regulation
14.
Pathol Res Pract ; 217: 153284, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33249400

ABSTRACT

OBJECTIVE: Lung adenocarcinoma (LUAD) is one of the most frequently occurring human malignancies worldwide, but its potential molecular mechanism has not yet been fully elucidated. N6-methyladenosine (m6A), the most common internal chemical modification of mRNAs, is implicated in diverse pathological processes in different human malignancies, but its functions in LUAD remain elusive. The current study aimed to investigate the function and molecular mechanism of KIAA1429 in LUAD. METHODS: The KIAA1429 expression level in LUAD tissues was assessed using databases and was detected in LUAD cells and tissues via quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blot. m6A levels in LUAD tissues and cells were quantified. Next, correlation between the KIAA1429 expression level and the clinical and pathological features and prognosis of patients with LUAD was analyzed. Further, KIAA1429 levels were decreased, and LUAD cell proliferation, migration, invasion, and cycle were assessed. Prediction websites revealed the aberrant expression and probable methylation modification of MUC3A in LUAD, and correlation between MUC3A and KIAA1429 was analyzed. Ultimately, the impact of the KIAA1429 expression on MUC3A-mediated malignant phenotypes of LUAD was examined by a torsion test. RESULTS: KIAA1429 expression was remarkably high and m6A level was aberrantly elevated in LUAD cells and tissues. In addition, high KIAA1429 expression indicated a larger tumor diameter, higher tumor-node-metastasis stage, greater proneness to lymph node and distant metastasis, and lower overall survival rate. siRNA-triggered KIAA1429 downregulation dramatically suppressed LUAD cell proliferation, migration, invasion, and cell cycle arrest in the G1 phase. Bioinformatics analysis revealed that MUC3A was expressed in LUAD at an unusually high level and may be methylated under the control of KIAA1429. Western blot, qRT-PCR, and correlation analyses revealed a positive correlation between KIAA1429 expression level and MUC3A. Finally, torsion test results revealed that low KIAA1429 expression reversed LUAD cell migration, proliferation, and invasion facilitated by low MUC3A expression as well as cell cycle arrest in the G1 phase. CONCLUSION: KIAA1429 exhibited an unusually high expression in LUAD cells and tissues, and high KIAA1429 expression was correlated with the clinical and pathological features of patients with LUAD, thereby leading to an unsatisfactory prognosis. Furthermore, KIAA1429 regulates MUC3A expression through m6A modification to modulate LUAD cells to proliferate, migrate, invade, and induce cell cycle arrest.


Subject(s)
Adenocarcinoma of Lung/metabolism , Adenosine/analogs & derivatives , Lung Neoplasms/metabolism , Mucin-3/metabolism , RNA Processing, Post-Transcriptional , RNA-Binding Proteins/metabolism , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/secondary , Adenosine/metabolism , Cell Movement , Cell Proliferation , Databases, Genetic , Female , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lymphatic Metastasis , Male , Methylation , Middle Aged , Mucin-3/genetics , Neoplasm Invasiveness , Neoplasm Staging , RNA-Binding Proteins/genetics , Signal Transduction , Tumor Burden
15.
Nutrients ; 12(2)2020 Feb 11.
Article in English | MEDLINE | ID: mdl-32053963

ABSTRACT

Amazake is a traditional Japanese beverage. Its main ingredients are sake cake and rice malt. In this study, we examined the effect of sake cake and rice malt on the intestinal barrier function and gut microbiota. BALB/c mice were fed a control diet or a diet containing a mixture of sake cake and rice malt powder (SRP) for four weeks. Fecal IgA values did not change between groups, but the fecal mucin level was significantly greater in the SRP-fed group. Gene expression analysis in the ileum by real-time PCR demonstrated Muc2 expression did not change, while the Muc3 expression was upregulated in the SRP-fed group. Furthermore, microbiota analysis demonstrated a change by SRP intake at the family level, and the proportion of Lactobacillaceae significantly increased in the SRP-fed group. At the genus level, the proportion of Lactobacillus also significantly increased in the SRP-fed group. These results suggest that the intake of a mixture of sake cake and rice malt improves intestinal barrier function by increasing mucin levels and inducing changes in intestinal microbiota.


Subject(s)
Animal Nutritional Physiological Phenomena , Beverages , Diet , Gastrointestinal Microbiome , Intestinal Mucosa/metabolism , Mucins/metabolism , Oryza , Animals , Feces/chemistry , Gene Expression , Ileum/metabolism , Lactobacillaceae , Male , Mice, Inbred BALB C , Mucin-3/genetics , Mucin-3/metabolism , Up-Regulation
16.
PLoS One ; 15(12): e0238540, 2020.
Article in English | MEDLINE | ID: mdl-33347507

ABSTRACT

BACKGROUND: Autoimmune hepatitis (AIH) is a disorder of unknown etiology in which immune-mediated liver injury progress to cirrhosis or hepatocellular carcinoma (HCC). The aim of the present study was to determine whether circulating soluble TIM3 (sTIM3) is elevated in patients with AIH patients and whether sTIM-3 levels are associated with clinical parameters of AIH. METHODS: We enrolled 123 Japanese patients with AIH who were identified from the National Hospital Organization-AIH-liver-network database, as well as 32 patients with chronic hepatitis C (CHC), 30 patients with primary biliary cholangitis (PBC) and healthy control subjects. Serum sTIM-3 concentrations were quantified by ELISA. RESULTS: Serum levels of sTIM-3 were significantly higher in AIH patients (median 4865 pg/ml; [interquartile range (IQR); 3122-7471]) compared to those in CHC (1026 pg/ml [IQR: 806-1283] p<0.001), PBC (2395 pg/ml [IQR: 2012-3422] p<0.001) or healthy controls (1285 pg/ml [IQR: 1098-1812] p<0.001). In AIH group, serum sTIM-3 were correlated with alanine aminotransferase (ALT), or total bilirubin (TB) and negatively correlated with serum levels of albumin (Alb). Serum levels of sTIM-3 were also strongly correlated with Mac-2 binding protein glycosylation isomer (M2BPGi) levels, but did not correlate with the histological grade of liver fibrosis. Steroid treatment of AIH patients significantly reduced serum sTIM-3 levels (2147±623pg/ml versus 1321±378pg/ml, p<0.001). CONCLUSIONS: Circulating sTIM-3 levels were elevated in AIH patients and are associated with AIH disease activity and AIH-related liver damage. These findings indicate that serum sTIM-3 correlated with disease status of AIH and could be useful biomarkers to detect autoimmune-mediated liver injury. Our data suggest a possible link between the TIM-3/GAL-9 pathway and AIH severity or phenotype, and further investigations of the TIM-3 pathway and AIH pathophysiology is warranted.


Subject(s)
Galectin 3/metabolism , Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/metabolism , Immunoglobulin Domains/immunology , Liver/immunology , Mucin-3/metabolism , T-Lymphocytes/immunology , Aged , Alanine Transaminase/immunology , Albumins/metabolism , Bilirubin/metabolism , Female , Glycosylation , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/metabolism , Humans , Liver/metabolism , Male , Middle Aged , T-Lymphocytes/metabolism
17.
Sci Rep ; 10(1): 16060, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32994477

ABSTRACT

During chronic hepatitis C virus (HCV) infection, both CD4+ and CD8+ T-cells become functionally exhausted, which is reflected by increased expression of programmed cell death-1 (PD-1) and T-cell immunoglobulin and mucin domain-containing protein 3 (Tim-3), and elevated anti-inflammatory interleukin 10 (IL-10) plasma levels. We studied 76 DAA-treated HCV-positive patients and 18 non-infected controls. Flow cytometry measured pretreatment frequencies of CD4+PD-1+, CD4+PD-1+Tim-3+ and CD8+PD-1+Tim-3+ T-cells and IL-10 levels measured by ELISA were significantly higher and CD4+PD-1-Tim-3- and CD8+PD-1-Tim-3- T-cells were significantly lower in patients than in controls. Treatment resulted in significant decrease of CD4+Tim-3+, CD8+Tim-3+, CD4+PD-1+Tim-3+ and CD8+PD-1+Tim-3+ T-cell frequencies as well as IL-10 levels and increase in CD4+PD-1-Tim-3- and CD8+PD-1-Tim-3- T-cells. There were no significant changes in the frequencies of CD4+PD-1+ T-cells, while CD8+PD-1+ T-cells increased. Patients with advanced liver fibrosis had higher PD-1 and lower Tim-3 expression on CD4+T-cells and treatment had little or no effect on the exhaustion markers. HCV-specific CD8+T-cells frequency has declined significantly after treatment, but their PD-1 and Tim-3 expression did not change. Successful treatment of chronic hepatitis C with DAA is associated with reversal of immune exhaustion phenotype, but this effect is absent in patients with advanced liver fibrosis.


Subject(s)
Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/pathology , Adult , Antiviral Agents/metabolism , Apoptosis , Biomarkers, Pharmacological/blood , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Female , Flow Cytometry , Hepatitis A Virus Cellular Receptor 2/immunology , Hepatitis C, Chronic/therapy , Humans , Interleukin-10/metabolism , Liver Cirrhosis/pathology , Male , Middle Aged , Mucin-3/metabolism , Plasma/metabolism , Programmed Cell Death 1 Receptor/metabolism
18.
Glycobiology ; 19(10): 1094-102, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19561031

ABSTRACT

A prominent feature of the rodent Muc3 SEA module is the precursor cleavage event that segregates the O-glycosylated N-terminal fragment and transmembrane domain into the noncovalently attached heterodimer. There are seven potential N-glycosylation sites that occur in a cluster in the SEA module of Muc3. However, it is unknown if these sites are modified or what the function of these N-glycans may be in the SEA module. Our data show that the proteolytic cleavage of the rodent Muc3 SEA module was partially prevented by treatment with tunicamycin, an inhibitor of N-glycosylation. Each single mutant of the seven N-glycosylation sites (N1A, N2A, N3A, N4A, N5A, N6A, and N7A) and multiple mutants, including double (N34A) and triple (N345A) mutants, and mutants with four (N3457A), five (N34567A), six (N134567A and N234567A), seven (N1234567A) mutations, confirmed that all seven of these potential sites are N-glycosylated simultaneously. The proteolytic cleavage of the SEA module was not affected when it lacked only one, two, or three N-glycans, but was partially inhibited when lacking four, five, and six N-glycans. In all, 2%, 48%, 85%, and 73% of the products from N3457A, N34567A, N134567A, and N234567A transfectants, respectively, remained uncleaved. The proteolytic cleavage was completely prevented in the N1234567A transfectant, which eliminated all seven N-glycans in the SEA module. The interaction of the heterodimer was independent of the N-glycans within the rodent Muc3 SEA module. Thus, the N-glycosylation pattern constituted a control point for the modulation of the proteolytic cleavage of the SEA module.


Subject(s)
Mucin-3/metabolism , Protein Processing, Post-Translational , Amino Acid Motifs , Animals , COS Cells , Chlorocebus aethiops , Conserved Sequence , Glycosylation/drug effects , Humans , Molecular Sequence Data , Mucin-3/chemistry , Mucin-3/genetics , Mutation/genetics , Rats , Sequence Alignment , Tunicamycin/pharmacology
19.
J Nutr ; 139(9): 1640-7, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19605529

ABSTRACT

We examined the role of soluble fiber viscosity in small intestinal mucin secretion. Viscosities were defined as the area under the viscosity curve (VAUC). Rats were fed a control diet or diets containing konjac mannan (KM) [low, medium, or high molecular weight (LKM, MKM, HKM), respectively] at 50 g/kg diet for 10 d. Luminal mucin content and goblet cell number increased in proportion to the molecular weight of KM. Such effects with the HKM diet were nullified by the concurrent ingestion of 2 g cellulase/kg diet. Diet containing LKM, MKM, HKM, guar gums (high or low molecular weight; HGG, LGG), psyllium (PS), or pectin (PC) at 50 g/kg was fed to rats. Fibers with higher VAUC (MKM, HKM, HGG, and PS) increased goblet cell numbers, but not those with lower VAUC (LKM, LGG, and PC). Luminal mucins were greater in rats fed HKM, PC, and PS diets. Goblet cell numbers and VAUC were correlated (r = 0.98; P < 0.01). In rats fed the HKM diet, ileal Muc2 gene expression was not affected, but that of Muc3 was lower than in those fed the control diet, indicating that the increase in luminal mucins after ingestion of HKM diet occurred independently of enhanced Muc gene expression. An incorporation study of 5'-bromo-deoxyuridine (BrdU) showed the position of the uppermost-BrdU labeled cell along the villi was higher in rats fed the HKM diet than in those fed the control diet. The results suggest that soluble fibers, except PC, upregulate baseline secretion of luminal mucins by increasing goblet cell numbers in proportion to fiber VAUC.


Subject(s)
Dietary Carbohydrates/pharmacology , Dietary Fiber/pharmacology , Gene Expression/drug effects , Goblet Cells/drug effects , Ileum/drug effects , Intestinal Mucosa/drug effects , Mucins/metabolism , Amorphophallus , Animals , Bromodeoxyuridine/metabolism , Cellulase/pharmacology , Diet , Epithelial Cells/drug effects , Galactans/pharmacology , Ileum/metabolism , Intestinal Mucosa/metabolism , Male , Mannans/pharmacology , Mucin-3/genetics , Mucin-3/metabolism , Mucins/genetics , Pectins/pharmacology , Plant Gums/pharmacology , Polystyrenes/pharmacology , Psyllium/pharmacology , Rats , Rats, Wistar , Solubility , Viscosity
20.
J Gastroenterol Hepatol ; 24(5): 920-6, 2009 May.
Article in English | MEDLINE | ID: mdl-19032457

ABSTRACT

BACKGROUND AND AIM: Chronic proliferative cholangitis (CPC) is currently considered as a pathological basis and major cause for the high recurrence rate of intrahepatic stones. Since CPC is a form of chronic proliferative disease, this study was designed to preliminarily investigate the inhibitory effect of proliferating cell nuclear antigen (PCNA) shRNA on the hyperplastic behavior and lithogenic potentiality of CPC. METHODS: The rat model of CPC was given an intralumenal administration of 0.5 mL PCNA shRNA through a 20-gauge venous retained needle. PCNA shRNA-mediated effects on CPC-associated hyperplastic behavior and lithogenic potential were assessed by investigating histological changes, immunohistochemistry for Ki-67, biochemistry for beta-glucuronidase, real-time polymerase chain reaction, and western blot analysis of PCNA, procollagen I, and mucin-3. RESULTS: PCNA shRNA treatment could efficiently inhibit the mRNA and protein expressions of the proliferation-related gene, PCNA, and Ki-67, which efficiently inhibited the hyperplastic behavior of the biliary epithelium, submucosal gland, and collagen fibers in the diseased bile duct wall. This novel treatment could efficiently inhibit the formation of acidic mucus glands, the expression of mucin-3 mRNA, and the secretion of endogenous beta-glucuronidase, thus effectively inhibiting the lithogenic potentiality of CPC. A further analysis revealed that PCNA shRNA-1 might display a more robust inhibitory effect on CPC-associated hyperplastic behavior and lithogenic potential than other gene sequences targeted in this study. CONCLUSIONS: PCNA shRNA-1 treatment could effectively inhibit the hyperplastic behavior and lithogenic potentiality of CPC, which might facilitate the prevention of stone recurrence and biliary restenosis.


Subject(s)
Cell Proliferation , Cholangitis/therapy , Genetic Therapy/methods , Proliferating Cell Nuclear Antigen/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Animals , Cholangitis/genetics , Cholangitis/metabolism , Cholangitis/pathology , Chronic Disease , Collagen Type I/metabolism , Disease Models, Animal , Glucuronidase/metabolism , Hyperplasia , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mucin-3/genetics , Mucin-3/metabolism , Proliferating Cell Nuclear Antigen/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/administration & dosage , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL