Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 57
1.
Cells ; 13(8)2024 Apr 14.
Article En | MEDLINE | ID: mdl-38667293

Chitinase 3-like 1 (also known as CHI3L1 or YKL-40) is a mammalian chitinase that has no enzymatic activity, but has the ability to bind to chitin, the polymer of N-acetylglucosamine (GlcNAc). Chitin is a component of fungi, crustaceans, arthropods including insects and mites, and parasites, but it is completely absent from mammals, including humans and mice. In general, chitin-containing organisms produce mammalian chitinases, such as CHI3L1, to protect the body from exogenous pathogens as well as hostile environments, and it was thought that it had a similar effect in mammals. However, recent studies have revealed that CHI3L1 plays a pathophysiological role by inducing anti-apoptotic activity in epithelial cells and macrophages. Under chronic inflammatory conditions such as inflammatory bowel disease and chronic obstructive pulmonary disease, many groups already confirmed that the expression of CHI3L1 is significantly induced on the apical side of epithelial cells, and activates many downstream pathways involved in inflammation and carcinogenesis. In this review article, we summarize the expression of CHI3L1 under chronic inflammatory conditions in various disorders and discuss the potential roles of CHI3L1 in those disorders on various cell types.


Chitinase-3-Like Protein 1 , Inflammation , Humans , Chitinase-3-Like Protein 1/metabolism , Animals , Inflammation/pathology , Inflammation/metabolism , Chronic Disease
2.
Oncol Lett ; 22(2): 618, 2021 Aug.
Article En | MEDLINE | ID: mdl-34257726

The tumor immune response is dependent on the interaction between tumor cells and the T-cell subset expressing the T-cell receptor (TCR) repertoire that infiltrates into the tumor microenvironment. The present study explored the diversity and shared TCR repertoires expressed on the surface of locoregional T cells and identified the T lymphocyte subsets infiltrating into esophageal squamous cell carcinoma (ESCC), in order to provide insight into the efficiency of immunotherapy and the development of a novel immune-oriented therapeutic strategy. A total of 53 patients with ESCC were enrolled in the present study, and immunohistochemical analysis of CD3, CD8, CD45RO, FOXP3, CD274, HLA class I and AE1/AE3 was performed. Digital pathological assessment was performed to evaluate the expression level of each marker. The clinicopathological significance of the immuno relation high (IR-Hi) group was assessed. Adaptor ligation PCR and next-generation sequencing were performed to explore the diversity of the TCR repertoire and to investigate the shared TCR repertoire in the IR-Hi group. Repertoire dissimilarity index (RDI) analysis was performed to assess the diversity of TCR, and the existence of shared TCRα and TCRß was also investigated. Further stratification was performed according to the expression of markers of different T-cell subsets. Patients were stratified into IR-Hi and immuno relation low (IR-Lo) groups. Cancer-specific survival and recurrence-free survival rates were significantly improved in the IR-Hi group compared with in the IT-Lo group. The diversity of the TCR repertoire was significantly higher in the IR-Hi group. TCR repertoire analysis revealed 27 combinations of TCRα and 23 combinations of TCRß VJ regions that were shared among the IR-Hi group. The IR-Hi group was divided into three clusters. Overall, the current findings revealed that the IR-Hi group maintained the diversity of TCR, and a portion of the IR-Hi cases held the T cells with shared TCR repertoires, implying recognition of shared antigens. The prognosis of patients with ESCC was affected by the existence of immune response cells and may possibly be stratified by the T-cell subsets.

3.
Diagnostics (Basel) ; 11(2)2021 Jan 30.
Article En | MEDLINE | ID: mdl-33573291

Inflammatory bowel disease (IBD) is a dysregulated inflammatory condition induced by multiple factors. The etiology of IBD is largely unknown, and the disease progression and prognosis are variable and unpredictable with uncontrolled disease behavior. Monitoring the status of chronic colitis closely is challenging for physicians, because the assessment of disease activity and severity require invasive methods. Using laboratory biomarkers may provide a useful alternative to invasive methods in the diagnosis and management of IBD. Furthermore, patients with ulcerative colitis or Crohn's disease are also at risk of developing cancer. Annual colonoscopies can help lower the risk for developing colorectal cancer. However, laboratory biomarkers may also be helpful as non-invasive indicators in predicting treatment responses, improving prognosis, and predicting possible tumors. This review addresses selected laboratory biomarkers (including ANCA, chitinase 3-like 1, S100A12/RAGE, calprotectin, and TNF/TNFR2), which are identified by utilizing two well-accepted animal models of colitis, dextran sodium sulfate-induced and T cell receptor alpha knockout colitis models. In addition to being useful for monitoring disease severity, these biomarkers are associated with therapeutic strategies. The factors may regulate the initiation and perpetuation of inflammatory factors in the gut.

4.
Cytokine ; 136: 155264, 2020 12.
Article En | MEDLINE | ID: mdl-32920320

BACKGROUND AND AIM: Interleukin-22 (IL-22), plays a vital role in the mucosal repair of inflammatory bowel disease (IBD). Serum levels of IL-22 and IL-22 binding protein (IL-22BP), a soluble inhibitory IL-22 receptor, were measured in patients with IBD to investigate the profile of IL-22 in the systemic circulation. METHODS: Blood samples from 92 healthy subjects, 98 patients with ulcerative colitis (UC), and 105 patients with Crohn's disease (CD) were analyzed for serum levels of IL-22, IL-22BP, human ß-defensin 2 (hBD-2), and serum inflammatory parameters. Disease activity was assessed by the partial Mayo score and Harvey-Bradshaw index for UC and CD, respectively. RESULTS: Serum IL-22 level was lower in UC (P < 0.001) and CD (P < 0.001) vs control and its decrease was more pronounced in CD than in UC (P = 0.019). Serum IL-22BP level was lower in UC (P < 0.001) and CD (P < 0.001) vs control and correlated with inflammatory parameters (albumin and C-reactive protein (CRP) in UC; hemoglobin, albumin, and CRP in CD). Serum IL-22/IL-22BP ratios were higher in UC (P = 0.009) vs control and correlated with inflammatory parameters (albumin and CRP). Serum hBD-2 level was higher only in CD (P = 0.015) but did not correlate with serum IL-22 levels, IL-22BP levels, IL-22/IL-22BP ratios, or inflammatory parameters. CONCLUSIONS: Dysregulation of the IL-22 system in the blood may play a role in the pathogenesis of IBD. Further studies are needed to understand the pathogenic and clinical significance of the blood IL-22 system in IBD.


Colitis, Ulcerative/blood , Crohn Disease/blood , Interleukins/blood , Adult , Biomarkers/blood , Female , Humans , Male , Middle Aged , Interleukin-22
5.
J Gastroenterol ; 53(4): 465-474, 2018 Apr.
Article En | MEDLINE | ID: mdl-29075900

IL-22 is a relatively new cytokine that is characterized by several unique biological properties. In the intestines, the effect of IL-22 is restricted mainly to non-lymphoid cells such as epithelial cells. Interestingly, the expression pattern and major cellular source of IL-22 have distinct difference between large and small intestines. IL-22 possesses an ability to constitutively activate STAT3 for promoting epithelial cell regeneration and reinforcing mucosal barrier integrity through stimulating the expression of anti-bacterial peptide and mucins. Of note, IL-22 is characterized as a two-faced cytokine that can play not only protective but also deleterious roles in the intestinal inflammation depending on the cytokine environment such as the expression levels of IL-23, T-bet, and IL-22 binding protein. Most importantly, clinical relevance of IL-22 to inflammatory bowel disease has been well highlighted. Mucosal healing, which represents the current therapeutic goal for IBD, can be induced by IL-22. Indeed, indigo naturalis, which can activate IL-22 pathway through Ahr, has been shown in a clinical trial to exhibit a strong therapeutic effect on ulcerative colitis. Despite the beneficial effect of IL-22, continuous activation of the IL-22 pathway increases the risk of colitis-associated cancer, particularly in patients with an extended history of IBD. This review article discusses how IL-22 regulates colitis, how beneficial versus deleterious effects of IL-22 is determined, and why IL-22 represents a promising target for IBD therapy.


Inflammatory Bowel Diseases/immunology , Interleukins/immunology , Gastrointestinal Agents/therapeutic use , Genetic Predisposition to Disease , Humans , Immunity, Mucosal , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Intestine, Large/immunology , Intestine, Small/immunology , Molecular Targeted Therapy/methods , Mucus/immunology , STAT3 Transcription Factor/metabolism , Wound Healing/immunology , Interleukin-22
6.
Proc Natl Acad Sci U S A ; 114(14): E2862-E2871, 2017 04 04.
Article En | MEDLINE | ID: mdl-28330995

The neonatal crystallizable fragment receptor (FcRn) is responsible for maintaining the long half-life and high levels of the two most abundant circulating proteins, albumin and IgG. In the latter case, the protective mechanism derives from FcRn binding to IgG in the weakly acidic environment contained within endosomes of hematopoietic and parenchymal cells, whereupon IgG is diverted from degradation in lysosomes and is recycled. The cellular location and mechanism by which FcRn protects albumin are partially understood. Here we demonstrate that mice with global or liver-specific FcRn deletion exhibit hypoalbuminemia, albumin loss into the bile, and increased albumin levels in the hepatocyte. In vitro models with polarized cells illustrate that FcRn mediates basal recycling and bidirectional transcytosis of albumin and uniquely determines the physiologic release of newly synthesized albumin into the basal milieu. These properties allow hepatic FcRn to mediate albumin delivery and maintenance in the circulation, but they also enhance sensitivity to the albumin-bound hepatotoxin, acetaminophen (APAP). As such, global or liver-specific deletion of FcRn results in resistance to APAP-induced liver injury through increased albumin loss into the bile and increased intracellular albumin scavenging of reactive oxygen species. Further, protection from injury is achieved by pharmacologic blockade of FcRn-albumin interactions with monoclonal antibodies or peptide mimetics, which cause hypoalbuminemia, biliary loss of albumin, and increased intracellular accumulation of albumin in the hepatocyte. Together, these studies demonstrate that the main function of hepatic FcRn is to direct albumin into the circulation, thereby also increasing hepatocyte sensitivity to toxicity.


Albumins/metabolism , Chemical and Drug Induced Liver Injury/genetics , Histocompatibility Antigens Class I/metabolism , Receptors, Fc/metabolism , Acetaminophen/adverse effects , Acetaminophen/metabolism , Animals , Bile/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Dogs , Female , Hepatocytes/metabolism , Histocompatibility Antigens Class I/genetics , Homeostasis , Madin Darby Canine Kidney Cells , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Receptors, Fc/genetics , Serum Albumin, Human/genetics , Serum Albumin, Human/metabolism , Transcytosis/genetics
8.
Inflamm Bowel Dis ; 22(5): 1137-50, 2016 May.
Article En | MEDLINE | ID: mdl-27070911

Inflammatory bowel disease (IBD) is an intestinal inflammatory condition that affects more than 2 million people in the United States. Although the etiology and pathogenesis of IBD are still largely unknown, dysregulated host/enteric microbial interactions are requisite for the development of IBD. So far, many researchers have tried to identify a precise relationship between IBD and an imbalance of the intestinal microbiota, termed "dysbiosis." Despite extensive efforts, it is still largely unknown about the interplay among microbes, their hosts, and their environments, and whether dysbiosis is a causal factor or an effect of IBD. Recently, deep-sequencing analyses of the microbiota in patients with IBD patients have been instrumental in characterizing the strong association between dysbiosis and IBD development, although it is still unable to identify specific-associated species level changes in most cases. Based on many recent reports, dysbiosis of the commensal microbiota is implicated in the pathogenesis of several diseases, including IBD, obesity, and allergic disorders, in both human and animal models. In this review article, the authors have focused on explaining the multiple types of dysbiosis, as well as dysbiosis-related diseases and potential treatments to apply this knowledge to understand a possible cause and potentially find therapeutic strategies for IBD as well as the other dysbiosis-related diseases.


Dysbiosis/epidemiology , Dysbiosis/pathology , Inflammatory Bowel Diseases/physiopathology , Models, Biological , Animals , Humans , Inflammatory Bowel Diseases/microbiology
9.
Cancer Res Front ; 2(1): 1-21, 2016 Feb.
Article En | MEDLINE | ID: mdl-27110580

To date, substantial evidence has shown a significant association between inflammatory bowel diseases (IBD) and development of colitis-associated cancer (CAC). The incidence/prevalence of IBD is higher in western countries including the US, Australia, and the UK. Although CAC development is generally characterized by stepwise accumulation of genetic as well as epigenetic changes, precise mechanisms of how chronic inflammation leads to the development of CAC are largely unknown. Preceding intestinal inflammation is one of the major influential factors for CAC tumorigenesis. Mucosal immune responses including activation of aberrant signaling pathways both in innate and adaptive immune cells play a pivotal role in CAC. Tumor progression and metastasis are shaped by a tightly controlled tumor microenvironment which is orchestrated by several immune cells and stromal cells including macrophages, neutrophils, dendritic cells, myeloid derived suppressor cells, T cells, and myofibroblasts. In this article, we will discuss the contributing factors of epithelial as well as immune cell signaling in initiation of CAC tumorigenesis and mucosal immune regulatory factors in the colonic tumor microenvironment. In depth understanding of these factors is necessary to develop novel anti-inflammatory and anti-cancer therapies for CAC in the near future.

10.
J Immunol ; 196(5): 2368-76, 2016 Mar 01.
Article En | MEDLINE | ID: mdl-26792803

The protein kinase p38α mediates cellular responses to environmental and endogenous cues that direct tissue homeostasis and immune responses. Studies of mice lacking p38α in several different cell types have demonstrated that p38α signaling is essential to maintaining the proliferation-differentiation balance in developing and steady-state tissues. The mechanisms underlying these roles involve cell-autonomous control of signaling and gene expression by p38α. In this study, we show that p38α regulates gut-associated lymphoid tissue (GALT) formation in a noncell-autonomous manner. From an investigation of mice with intestinal epithelial cell-specific deletion of the p38α gene, we find that p38α serves to limit NF-κB signaling and thereby attenuate GALT-promoting chemokine expression in the intestinal epithelium. Loss of this regulation results in GALT hyperplasia and, in some animals, mucosa-associated B cell lymphoma. These anomalies occur independently of luminal microbial stimuli and are most likely driven by direct epithelial-lymphoid interactions. Our study illustrates a novel p38α-dependent mechanism preventing excessive generation of epithelial-derived signals that drive lymphoid tissue overgrowth and malignancy.


Intestinal Mucosa/metabolism , Mitogen-Activated Protein Kinase 14/metabolism , NF-kappa B/metabolism , Peyer's Patches/immunology , Peyer's Patches/metabolism , Signal Transduction , Animals , Cell Line , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/microbiology , Colon/pathology , Epithelial Cells/metabolism , Gene Expression , Hyperplasia , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Knockout , Mice, Transgenic , Microbiota/immunology , Mitogen-Activated Protein Kinase 14/genetics , Peyer's Patches/pathology
11.
J Pathol ; 238(2): 205-19, 2016 Jan.
Article En | MEDLINE | ID: mdl-26387641

Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition that is mediated by very complex mechanisms controlled by genetic, immune, and environmental factors. More than 74 kinds of genetically engineered mouse strains have been established since 1993 for studying IBD. Although mouse models cannot fully reflect human IBD, they have provided significant contributions for not only understanding the mechanism, but also developing new therapeutic means for IBD. Indeed, 20 kinds of genetically engineered mouse models carry the susceptibility genes identified in human IBD, and the functions of some other IBD susceptibility genes have also been dissected out using mouse models. Cutting-edge technologies such as cell-specific and inducible knockout systems, which were recently employed to mouse IBD models, have further enhanced the ability of investigators to provide important and unexpected rationales for developing new therapeutic strategies for IBD. In this review article, we briefly introduce 74 kinds of genetically engineered mouse models that spontaneously develop intestinal inflammation.


Disease Models, Animal , Genetic Engineering , Inflammatory Bowel Diseases/genetics , Animals , Cytokines/genetics , Gene Targeting , Genes, Neoplasm/genetics , Genetic Predisposition to Disease/genetics , Humans , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Signal Transduction/genetics
12.
PLoS One ; 10(10): e0139149, 2015.
Article En | MEDLINE | ID: mdl-26440614

Colorectal cancer (CRC) development is mediated by uncontrolled survival and proliferation of tumor progenitor cells. Using animal models to identify and study host-derived factors that underlie this process can aid interventions in preventing tumor expansion and metastasis. In healthy steady states in humans and mice (e.g. C57BL/6 strain), colonic Chitinase 3-like 1 (CHI3L1) gene expression is undetectable. However, this expression can be induced during intestinal inflammation and tumorigenesis where CHI3L1 plays an important role in tissue restitution and cell proliferation. Here, we show that a wild-derived mouse strain MOLF/EiJ expresses high levels of colonic epithelial CHI3L1 at the steady state due to several nucleotide polymorphisms in the proximal promoter regions of the CHI3L1 gene. Interestingly, these mice spontaneously developed polypoid nodules in the colon with signs of immune cell infiltrations at steady state. The CHI3L1 positive colonic epithelial cells were highly proliferative and exhibited malignant transformation and expansion when exposed in vivo to azoxymethane, one of the well-known colonic carcinogens.


Colonic Neoplasms/metabolism , Glycoproteins/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chitinase-3-Like Protein 1 , Colonic Neoplasms/genetics , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glycoproteins/genetics , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Confocal
13.
Oncotarget ; 6(34): 36535-50, 2015 Nov 03.
Article En | MEDLINE | ID: mdl-26431492

Many host-factors are inducibly expressed during the development of inflammatory bowel disease (IBD), each having their unique properties, such as immune activation, bacterial clearance, and tissue repair/remodeling. Dysregulation/imbalance of these factors may have pathogenic effects that can contribute to colitis-associated cancer (CAC). Previous reports showed that IBD patients inducibly express colonic chitinase 3-like 1 (CHI3L1) that is further upregulated during CAC development. However, little is known about the direct pathogenic involvement of CHI3L1 in vivo. Here we demonstrate that CHI3L1 (aka Brp39) knockout (KO) mice treated with azoxymethane (AOM)/dextran sulphate sodium (DSS) developed severe colitis but lesser incidence of CAC as compared to that in wild-type (WT) mice. Highest CHI3L1 expression was found during the chronic phase of colitis, rather than the acute phase, and is essential to promote intestinal epithelial cell (IEC) proliferation in vivo. This CHI3L1-mediated cell proliferation/survival involves partial downregulation of the pro-apoptotic S100A9 protein that is highly expressed during the acute phase of colitis, by binding to the S100A9 receptor, RAGE (Receptor for Advanced Glycation End products). This interaction disrupts the S100A9-associated expression positive feedback loop during early immune activation, creating a CHI3L1hi S100A9low colonic environment, especially in the later phase of colitis, which promotes cell proliferation/survival of both normal IECs and tumor cells.


Calgranulin B/metabolism , Chitinase-3-Like Protein 1/metabolism , Colonic Neoplasms/metabolism , Animals , Cell Proliferation/physiology , Chitinase-3-Like Protein 1/biosynthesis , Chitinase-3-Like Protein 1/genetics , Chronic Disease , Colitis/enzymology , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Disease Models, Animal , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , S100 Calcium Binding Protein beta Subunit/metabolism
14.
Methods Mol Biol ; 1207: 231-48, 2015.
Article En | MEDLINE | ID: mdl-25253144

The intestine, which provides the first line of defense against over trillion of enteric microorganisms, suffers from broad range of inflammatory conditions caused by infectious, autoimmune, allergic, neurological, and ischemic mechanisms. Recent data have suggested dual roles (protective versus deleterious) for galectins in the pathogenesis of some intestinal inflammations, highlighting the importance of this area of research. A potential problem with the research of intestinal inflammation may be the requirement of some unique techniques. Therefore, we herein describe how to induce intestinal inflammation and how to isolate lymphocyte, myeloid cell, follicular cell, and epithelial cell populations separately from the intestine for the study of intestinal inflammations.


Galectins/metabolism , Intestinal Diseases/metabolism , Intestinal Diseases/pathology , Animals , B-Lymphocytes/cytology , Cell Separation , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Female , Fibroblasts/pathology , Humans , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intestinal Diseases/immunology , Leukocyte Common Antigens/metabolism , Macrophages/cytology , Male , Mice , T-Lymphocytes/cytology
15.
Methods Mol Biol ; 1190: 227-41, 2014.
Article En | MEDLINE | ID: mdl-25015284

Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition with increasing incidence and prevalence around the world. Although B cells had generally been believed to play a pathogenic role in IBD due to the production of autoantibodies, a growing body of evidence from mouse models suggests the coexistence of pathogenic B cells and regulatory B cells, termed Breg, in this disorder. Since some unique techniques are required to closely study the Breg in gut-associated lymphoid tissues (GALT), we herein describe how to induce colitis in mice and how to analyze the phenotype and function of GALT-specific Breg.


B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/pathology , Colitis/immunology , Colon/pathology , Disease Models, Animal , Lymphoid Tissue/pathology , Animals , Cell Culture Techniques/methods , Cell Separation/methods , Colitis/chemically induced , Colitis/pathology , Colon/cytology , Colon/immunology , Cytokines/analysis , Cytokines/immunology , Dextran Sulfate , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Inbred C57BL
16.
Inflamm Bowel Dis ; 20(5): 835-46, 2014 May.
Article En | MEDLINE | ID: mdl-24694795

BACKGROUND: Chitinase 3-like 1 (CHI3L1) is an inducible molecule on intestinal epithelial cells during the development of inflammatory bowel disease. METHODS: To investigate the role of CHI3L1 in bacterial infectious colitis, we orally inoculated pathogenic Salmonella typhimurium and potentially pathogenic adherent-invasive Escherichia coli (AIEC) LF82 virulent strain into C57Bl/6 wild-type mice or CHI3L1 knockout (KO) mice. RESULTS: Both S. typhimurium and AIEC LF82 were found to efficiently induce severe intestinal inflammation in wild-type mice but not in CHI3L1 KO mice. These bacteria-infected CHI3L1 KO mice exhibit decreased cellular infiltration, bacterial translocation, and production of interleukin (IL)-6 and IL-22, as compared with those of wild-type mice. More importantly, CHI3L1 KO mice displayed aberrant STAT3 activation after bacterial infections. Co-stimulation of CHI3L1 and IL-6, but not IL-22, synergistically activates STAT3 signaling pathway in intestinal epithelial cells in an NF-κB/MAPK-dependent manner. CONCLUSIONS: CHI3L1 promotes the onset of selected gram-negative bacterial infectious colitis through IL-6/STAT3 pathway.


Colitis/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Glycoproteins/physiology , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , STAT3 Transcription Factor/metabolism , Animals , Bacterial Adhesion , Blotting, Western , Cells, Cultured , Chitinase-3-Like Protein 1 , Colitis/microbiology , Colitis/pathology , Epithelial Cells/microbiology , Epithelial Cells/pathology , Escherichia coli/pathogenicity , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/pathology , Immunoenzyme Techniques , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phosphorylation , Salmonella Infections, Animal/metabolism , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/pathogenicity , Signal Transduction
17.
World J Gastroenterol ; 19(32): 5238-49, 2013 Aug 28.
Article En | MEDLINE | ID: mdl-23983426

Inflammatory bowel disease (IBD) is a consequence of the complex, dysregulated interplay between genetic predisposition, environmental factors, and microbial composition in the intestine. Despite a great advancement in identifying host-susceptibility genes using genome-wide association studies (GWAS), the majority of IBD cases are still underrepresented. The immediate challenge in post-GWAS era is to identify other causative genetic factors of IBD. DNA methylation has received increasing attention for its mechanistical role in IBD pathogenesis. This stable, yet dynamic DNA modification, can directly affect gene expression that have important implications in IBD development. The alterations in DNA methylation associated with IBD are likely to outset as early as embryogenesis all the way until old-age. In this review, we will discuss the recent advancement in understanding how DNA methylation alterations can contribute to the development of IBD.


DNA Methylation , Epigenesis, Genetic , Inflammatory Bowel Diseases/genetics , Animals , DNA Methylation/drug effects , DNA, Bacterial/metabolism , Epigenesis, Genetic/drug effects , Gastrointestinal Agents/therapeutic use , Genetic Predisposition to Disease , Host-Pathogen Interactions , Humans , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Intestines/microbiology , Phenotype , Risk Factors
18.
J Biol Chem ; 288(33): 23788-97, 2013 Aug 16.
Article En | MEDLINE | ID: mdl-23836897

The epithelium of mucosal and skin surfaces serves as a permeability barrier and affords mechanisms for local immune defense. Crucial to the development and maintenance of a properly functioning epithelium is the balance of cell proliferation, differentiation, and death. Here we show that this balance depends on cross-regulatory interactions among multiple protein kinase-mediated signals and their coordinated transmission. From an investigation of conditional gene knock-out mice, we find that epithelial-specific loss of the protein kinase p38α leads to aberrant activation of TAK1, JNK, EGF receptor, and ERK in distinct microanatomical areas of the intestines and skin. Consequently, the epithelial tissues display excessive proliferation, inadequate differentiation, and sensitivity to apoptosis. These anomalies leave the tissue prone to damage and collapse at the trigger of an environmental insult. The vulnerability of p38α-deficient epithelium predicts adverse effects of long term pharmacological p38α inhibition; yet such limitations could be overcome by concomitant blockade of one or more of the dysregulated protein kinase signaling pathways.


Epithelium/enzymology , Homeostasis , MAP Kinase Signaling System , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Enzyme Activation , Epithelial Cells/enzymology , Epithelial Cells/pathology , Epithelium/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammation/enzymology , Inflammation/pathology , Intestinal Mucosa/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , Skin/pathology , Ubiquitination , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
19.
J Exp Med ; 209(13): 2383-94, 2012 Dec 17.
Article En | MEDLINE | ID: mdl-23209314

Immune responses are modified by a diverse and abundant repertoire of carbohydrate structures on the cell surface, which is known as the glycome. In this study, we propose that a unique glycome that can be identified through the binding of galectin-4 is created on local, but not systemic, memory CD4+ T cells under diverse intestinal inflammatory conditions, but not in the healthy state. The colitis-associated glycome (CAG) represents an immature core 1-expressing O-glycan. Development of CAG may be mediated by down-regulation of the expression of core-2 ß1,6-N-acetylglucosaminyltransferase (C2GnT) 1, a key enzyme responsible for the production of core-2 O-glycan branch through addition of N-acetylglucosamine (GlcNAc) to a core-1 O-glycan structure. Mechanistically, the CAG seems to contribute to super raft formation associated with the immunological synapse on colonic memory CD4+ T cells and to the consequent stabilization of protein kinase C θ activation, resulting in the stimulation of memory CD4+ T cell expansion in the inflamed intestine. Functionally, CAG-mediated CD4+ T cell expansion contributes to the exacerbation of T cell-mediated experimental intestinal inflammations. Therefore, the CAG may be an attractive therapeutic target to specifically suppress the expansion of effector memory CD4+ T cells in intestinal inflammation such as that seen in inflammatory bowel disease.


CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Immunologic Memory , Polysaccharides/immunology , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Disease Models, Animal , Down-Regulation , Enzyme Activation , Galectin 4/immunology , Galectin 4/metabolism , Humans , Immunological Synapses/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Isoenzymes/metabolism , Leukocyte Common Antigens/metabolism , Lymphocyte Activation , Membrane Microdomains/immunology , Membrane Microdomains/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Polysaccharides/metabolism , Protein Kinase C/metabolism , Protein Kinase C-theta
20.
Inflamm Bowel Dis ; 18(9): 1777-84, 2012 Sep.
Article En | MEDLINE | ID: mdl-22359410

An interleukin (IL)-10 family cytokine, IL-22 is characterized by several unique biological properties, including 1) the target restricted to innate cells; 2) the distinct expression pattern between large and small intestines; 3) alteration of the cellular source depending on several factors; 4) the dual abilities to serve as protective versus proinflammatory mediators in inflammatory responses; and 5) the close association with some major inflammatory bowel disease (IBD) susceptibility genes. The major functions of IL-22 in the intestine are the stimulation of epithelial cells to produce a wide variety of antibacterial proteins, the reinforcement of mucus barrier through stimulation of mucin 1 production under intestinal inflammatory conditions, and the enhancement of epithelial regeneration with goblet cell restitution. Through these beneficial functions, IL-22 contributes to the improvement of some types of experimental chronic colitis, which are mediated by T helper (Th)1 or Th2 responses. Most important, studies using both loss-of-function and gain-of-function approaches have clearly demonstrated the ability of IL-22 to promote intestinal wound healing from acute intestinal injury. These findings highlight IL-22 as an attractive and promising target for future IBD therapy. Alternatively, the enormous progress in the field of IL-22 biology has also suggested more complicated mechanisms with the IL-22 pathway than previously predicted. This review article briefly summarizes previous and current knowledge on IL-22 particularly associated with intestinal inflammation.


Inflammation/prevention & control , Interleukins/therapeutic use , Intestines/drug effects , Animals , Humans , Intestines/pathology , Interleukin-22
...