Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 10(3): 673-684, 2017 05.
Article in English | MEDLINE | ID: mdl-27624780

ABSTRACT

Specific components of the intestinal microbiota are capable of influencing immune responses such that a mutualistic relationship is established. In mice, colonization with segmented filamentous bacteria (SFB) induces T-helper-17 (Th17) cell differentiation in the intestine, yet the effector functions of interleukin (IL)-17A in response to SFB remain incompletely understood. Here we report that colonization of mice with SFB-containing microbiota induced IL-17A- and CXCR2-dependent recruitment of neutrophils to the ileum. This response required adaptive immunity, as Rag-deficient mice colonized with SFB-containing microbiota failed to induce IL-17A, CXCL1 and CXCL2, and displayed defective neutrophil recruitment to the ileum. Interestingly, neutrophil depletion in wild-type mice resulted in significantly augmented Th17 responses and SFB expansion, which correlated with impaired expression of IL-22 and antimicrobial peptides. These data provide novel insight into a dynamic IL-17A-CXCR2-neutrophil axis during acute SFB colonization and demonstrate a central role for neutrophils in limiting SFB expansion.


Subject(s)
Bacteria/immunology , Gastrointestinal Microbiome/immunology , Ileum/immunology , Interleukin-17/metabolism , Neutrophils/immunology , Receptors, Interleukin-8B/metabolism , Th17 Cells/immunology , Adaptive Immunity/genetics , Animals , Antimicrobial Cationic Peptides/metabolism , Bacteria/growth & development , Cell Differentiation , Cell Movement/genetics , Cells, Cultured , Homeodomain Proteins/genetics , Ileum/microbiology , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Interleukin-22
2.
Clin Exp Immunol ; 184(3): 347-57, 2016 06.
Article in English | MEDLINE | ID: mdl-26822517

ABSTRACT

Noroviruses (NoV) are the most common cause of epidemic gastroenteritis world-wide. NoV infections are often asymptomatic, although individuals still shed large amounts of NoV in their stool. Understanding the differences between asymptomatic and symptomatic individuals would help in elucidating mechanisms of NoV pathogenesis. Our goal was to compare the serum cytokine responses and faecal viral RNA titres of asymptomatic and symptomatic NoV-infected individuals. We tested serum samples from infected subjects (n = 26; 19 symptomatic, seven asymptomatic) from two human challenge studies of GI.1 NoV for 16 cytokines. Samples from prechallenge and days 1-4 post-challenge were tested for these cytokines. Cytokine levels were compared to stool NoV RNA titres quantified previously by reverse transcription-polymerase chain reaction (RT-qPCR). While both symptomatic and asymptomatic groups had similar patterns of cytokine responses, the symptomatic group generally exhibited a greater elevation of T helper type 1 (Th1) and Th2 cytokines and IL-8 post-challenge compared to the asymptomatic group (all P < 0·01). Daily viral RNA titre was associated positively with daily IL-6 concentration and negatively with daily IL-12p40 concentration (all P < 0·05). Symptoms were not associated significantly with daily viral RNA titre, duration of viral shedding or cumulative shedding. Symptomatic individuals, compared to asymptomatic, have greater immune system activation, as measured by serum cytokines, but they do not have greater viral burden, as measured by titre and shedding, suggesting that symptoms may be immune-mediated in NoV infection.


Subject(s)
Gastroenteritis/diagnosis , Interleukin-12 Subunit p40/blood , Interleukin-6/blood , Interleukin-8/blood , Norovirus/immunology , Virus Shedding/immunology , Adolescent , Adult , Asymptomatic Diseases , Feces/chemistry , Feces/virology , Female , Gastroenteritis/immunology , Gastroenteritis/pathology , Gastroenteritis/virology , Host-Pathogen Interactions , Humans , Immunity, Innate , Male , Norovirus/genetics , Norovirus/growth & development , RNA, Viral/genetics , RNA, Viral/immunology , Severity of Illness Index , Th1 Cells/immunology , Th1 Cells/pathology , Th1 Cells/virology , Th1-Th2 Balance , Th2 Cells/immunology , Th2 Cells/pathology , Th2 Cells/virology , Viral Load/immunology
3.
Mucosal Immunol ; 9(5): 1151-62, 2016 09.
Article in English | MEDLINE | ID: mdl-26732677

ABSTRACT

A characteristic feature of gastrointestinal tract inflammatory disorders, such as inflammatory bowel disease, is polymorphonuclear neutrophil (PMN) transepithelial migration (TEM) and accumulation in the gut lumen. PMN accumulation within the intestinal mucosa contributes to tissue injury. Although epithelial infiltration by large numbers of PMNs results in mucosal injury, we found that PMN interactions with luminal epithelial membrane receptors may also play a role in wound healing. Intercellular adhesion molecule-1 (ICAM-1) is a PMN ligand that is upregulated on apical surfaces of intestinal epithelial cells under inflammatory conditions. In our study, increased expression of ICAM-1 resulted in enhanced PMN binding to the apical epithelium, which was associated with reduced PMN apoptosis. Following TEM, PMN adhesion to ICAM-1 resulted in activation of Akt and ß-catenin signaling, increased epithelial-cell proliferation, and wound healing. Such responses were ICAM-1 dependent as engagement of epithelial ICAM-1 by antibody-mediated cross-linking yielded similar results. Furthermore, using an in-vivo biopsy-based, colonic-mucosal-injury model, we demonstrated epithelial ICAM-1 has an important role in activation of epithelial Akt and ß-catenin signaling and wound healing. These findings suggest that post-migrated PMNs within the intestinal lumen can regulate epithelial homeostasis, thereby identifying ICAM-1 as a potential therapeutic target for promoting mucosal wound healing.


Subject(s)
Colon/immunology , Epithelial Cells/immunology , Immunity, Mucosal , Intercellular Adhesion Molecule-1/immunology , Intestinal Mucosa/immunology , Neutrophils/immunology , Wound Healing/immunology , Animals , Biopsy , Cell Communication/immunology , Cell Line , Cell Proliferation , Colon/injuries , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Intercellular Adhesion Molecule-1/genetics , Intestinal Mucosa/injuries , Male , Mice , Mice, Inbred C57BL , Neutrophils/cytology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Signal Transduction , Tissue Culture Techniques , Transendothelial and Transepithelial Migration/immunology , beta Catenin/genetics , beta Catenin/immunology
4.
Clin Exp Immunol ; 182(2): 195-203, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26178578

ABSTRACT

Noroviruses (NoV) are the most common cause of epidemic gastroenteritis worldwide. The acute immune response to NoV in humans is poorly understood, hindering research on prevention and treatment. To elucidate the acute immune response and test for cytokine predictors of susceptibility to infection, serum samples from two human NoV challenge studies were tested for 16 cytokines. Subjects who became infected (n = 26) were age-matched with subjects who remained uninfected following NoV challenge (n = 26). Samples were tested from prechallenge and days 1-4 post-challenge. Cytokine responses were compared between infected and uninfected groups. Overall, infected individuals exhibited an elevation in T helper type 1 (Th1) and Th2 cytokines, as well as chemokines interleukin (IL)-8 and monocyte chemoattractant protein (MCP-1), compared to uninfected individuals (all P < 0.05). Most cytokines peaked on day 2 post-challenge in infected subjects, and tumour necrosis factor (TNF)-α, IL-8, and IL-10 remained elevated to day 3. The only cytokine elevated significantly among infected subjects to day 4 post-challenge was IL-10 (P = 0.021). Prechallenge cytokine concentrations were not predictive of infection status post-challenge. There were no significant changes in serum cytokines among NoV-challenged subjects who remained uninfected. These results suggest that NoV infection elicits a Th1-type response, with some Th2 activation. Persistent elevation of IL-10 among infected subjects is consistent with activation of adaptive immune responses, such as B cell expansion, as well as down-regulation of Th1 cytokines. This study presents the first comprehensive description of the acute cytokine response to GI.1 NoV in humans.


Subject(s)
Caliciviridae Infections/immunology , Cytokines/immunology , Gastroenteritis/immunology , Norovirus/immunology , Adult , Caliciviridae Infections/blood , Caliciviridae Infections/virology , Chemokine CCL2/blood , Chemokine CCL2/immunology , Cytokines/blood , Feces/virology , Female , Gastroenteritis/blood , Gastroenteritis/virology , Host-Pathogen Interactions/immunology , Humans , Interleukin-10/blood , Interleukin-10/immunology , Interleukin-8/blood , Interleukin-8/immunology , Male , Norovirus/genetics , Norovirus/physiology , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Time Factors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology , Young Adult
5.
Mucosal Immunol ; 7(5): 1221-32, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24621992

ABSTRACT

Neutrophil transepithelial migration (TEM) during acute inflammation is associated with mucosal injury. Using models of acute mucosal injury in vitro and in vivo, we describe a new mechanism by which neutrophils infiltrating the intestinal mucosa disrupt epithelial homeostasis. We report that junctional adhesion molecule-like protein (JAML) is cleaved from neutrophil surface by zinc metalloproteases during TEM. Neutrophil-derived soluble JAML binds to the epithelial tight junction protein coxsackie-adenovirus receptor (CAR) resulting in compromised barrier and inhibition of wound repair, through decreased epithelial proliferation. The deleterious effects of JAML on barrier and wound repair are reversed with an anti-JAML monoclonal antibody that inhibits JAML-CAR binding. JAML released from transmigrating neutrophils across inflamed epithelia may thus promote recruitment of leukocytes and aid in clearance of invading microorganisms. However, sustained release of JAML under pathologic conditions associated with persistence of large numbers of infiltrated neutrophils would compromise intestinal barrier and inhibit mucosal healing. Thus, targeting JAML-CAR interactions may improve mucosal healing responses under conditions of dysregulated neutrophil recruitment.


Subject(s)
Cell Adhesion Molecules/metabolism , Epithelial Cells/immunology , Inflammation/physiopathology , Intestinal Diseases/physiopathology , Neutrophils/immunology , Animals , Apoptosis , CHO Cells , Cell Adhesion Molecules/immunology , Cell Line , Cell Proliferation , Coxsackie and Adenovirus Receptor-Like Membrane Protein/metabolism , Cricetulus , Epithelial Cells/cytology , HL-60 Cells , Humans , Inflammation/immunology , Intestinal Diseases/immunology , Models, Immunological , Protein Binding , Wound Healing/immunology
6.
Oncogene ; 33(36): 4531-6, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-24166502

ABSTRACT

Desmosomal cadherins mediate cell-cell adhesion in epithelial tissues and have been known to be altered in cancer. We have previously shown that one of the two intestinal epithelial desmosomal cadherins, desmocollin-2 (Dsc2) loss promotes colonic epithelial carcinoma cell proliferation and tumor formation. In this study we show that loss of the other intestinal desmosomal cadherin, desmoglein-2 (Dsg2) that pairs with Dsc2, results in decreased epithelial cell proliferation and suppressed xenograft tumor growth in mice. Dsg2-deficient cells demonstrated a compensatory increase in Dsc2 expression, and small interfering RNA-mediated loss of Dsc2 restored proliferation in Dsg2-deficient cells. Dsg2 downregulation inhibited epidermal growth factor receptor (EGFR) signaling and cell proliferation through altered phosphorylation of EGFR and downstream extracellular signal-regulated kinase activation in parallel with inhibited EGFR receptor internalization. Additionally, we demonstrated a central role of Dsc2 in controlling EGFR signaling and cell proliferation in intestinal epithelial cells. Consistent with these findings, analyses of human colon cancers demonstrated increased Dsg2 protein expression. Taken together, these data demonstrate that partner desmosomal cadherins Dsg2 and Dsc2 play opposing roles in controlling colonic carcinoma cell proliferation through differential effects on EGFR signaling.


Subject(s)
Adenocarcinoma/pathology , Colonic Neoplasms/pathology , Desmocollins/metabolism , Desmogleins/metabolism , Adenocarcinoma/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/metabolism , Desmocollins/genetics , Desmogleins/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Male , Mice , Neoplasms, Experimental , Signal Transduction
7.
Mucosal Immunol ; 7(4): 905-15, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24345805

ABSTRACT

Neutrophil (PMN) transepithelial migration (TEM) and accumulation in luminal spaces is a hallmark of mucosal inflammation. TEM has been extensively modeled; however, the functional consequences and molecular basis of PMN interactions with luminal epithelial ligands are not clear. Here we report that cytokine-induced expression of a PMN ligand, intercellular adhesion molecule-1 (ICAM-1), exclusively on the luminal (apical) membrane of the intestinal epithelium results in accumulation and enhanced motility of transmigrated PMN on the apical epithelial surface. Using complementary in-vitro and in-vivo approaches, we demonstrate that ligation of epithelial ICAM-1 by PMN or with specific antibodies results in myosin light-chain kinase-dependent increases in epithelial permeability that are associated with enhanced PMN TEM. Effects of ICAM-1 ligation on epithelial permeability and PMN migration in vivo were blocked after intraluminal addition of peptides derived from the cytoplasmic domain of ICAM-1. These findings provide new evidence for functional interactions between PMN and epithelial cells after migration into the intestinal lumen. Although such interactions may aid in clearance of invading microorganisms by promoting PMN recruitment, engagement of ICAM-1 under pathologic conditions would increase accumulation of epithelial-associated PMN, thus contributing to mucosal injury as observed in conditions, including ulcerative colitis.


Subject(s)
Intercellular Adhesion Molecule-1/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Transendothelial and Transepithelial Migration/immunology , Animals , Antibodies/immunology , Antibodies/metabolism , Cell Adhesion/immunology , Cell Line , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Interferon-gamma/metabolism , Intestinal Mucosa/pathology , Male , Mice , Models, Biological , Myosin-Light-Chain Kinase/metabolism , Neutrophil Infiltration/immunology , Permeability , Transendothelial and Transepithelial Migration/genetics
8.
Mucosal Immunol ; 6(1): 136-45, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22763408

ABSTRACT

The molecular mechanisms that restore intestinal epithelial homeostasis during colitis are incompletely understood. Here, we report that during intestinal inflammation, multiple inflammatory cytokines promote the activity of a master regulator of cell proliferation and apoptosis, serine/threonine kinase CK2. Enhanced mucosal CK2 protein expression and activity were observed in animal models of chronic colitis, particularly within intestinal epithelial cells (IECs). The in vitro treatment of intestinal epithelial cell lines with cytokines resulted in increased CK2 expression and nuclear translocation of its catalytic α subunit. Similarly, nuclear translocation of CK2α was a prominent feature observed in colonic crypts from individuals with ulcerative colitis and Crohn's disease. Further in vitro studies revealed that CK2 activity promotes epithelial restitution, and protects normal IECs from cytokine-induced apoptosis. These observations identify CK2 as a key regulator of homeostatic properties of the intestinal epithelium that serves to promote wound healing, in part through inhibition of apoptosis under conditions of inflammation.


Subject(s)
Casein Kinase II/metabolism , Colitis/immunology , Colitis/metabolism , Homeostasis/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Animals , Apoptosis/genetics , Casein Kinase II/genetics , Caspases/metabolism , Cell Line , Cell Nucleus/metabolism , Cell Proliferation , Colitis/chemically induced , Colitis/genetics , Disease Models, Animal , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Mice , Protein Transport , Rats , Wound Healing , beta Catenin/metabolism
9.
Mucosal Immunol ; 5(6): 681-90, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22692453

ABSTRACT

Proinflammatory cytokines induce guanylate-binding protein 1 (GBP-1) protein expression in intestinal epithelial tissues. GBP-1 has been described as influencing a number of cellular processes important for epithelial homeostasis, including cell proliferation. However, many questions remain as to the role of GBP-1 in intestinal mucosal homeostasis. We therefore sought to investigate the function of proinflammatory cytokine-induced GBP-1 during intestinal epithelial cell proliferation. Through the use of complementary GBP-1 overexpression and small interfering RNA-mediated knockdown studies, we now show that GBP-1 acts to inhibit pro-mitogenic ß-catenin/T cell factor (TCF) signaling. Interestingly, proinflammatory cytokine-induced GBP-1 was found to be a potent suppressor of ß-catenin protein levels and ß-catenin serine 552 phosphorylation. Neither glycogen synthase kinase 3ß nor proteasomal inhibition alleviated GBP-1-mediated suppression of cell proliferation or ß-catenin/TCF signaling, indicating a non-canonical mechanism of ß-catenin inhibition. Together, these data show that cytokine-induced GBP-1 retards cell proliferation by forming a negative feedback loop that suppresses ß-catenin/TCF signaling.


Subject(s)
Epithelial Cells/metabolism , GTP-Binding Proteins/genetics , Interferon-gamma/pharmacology , TCF Transcription Factors/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , beta Catenin/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/immunology , Feedback, Physiological/drug effects , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/immunology , Gene Expression/drug effects , Gene Expression/immunology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/immunology , Glycogen Synthase Kinase 3 beta , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Serine/metabolism , Signal Transduction/drug effects , TCF Transcription Factors/genetics , TCF Transcription Factors/immunology , beta Catenin/genetics , beta Catenin/immunology
10.
Mucosal Immunol ; 5(4): 354-66, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22491176

ABSTRACT

Polymorphonuclear leukocytes or neutrophils play a critical role in the maintenance of intestinal homeostasis. They have elegant defense mechanisms to eliminate microbes that have translocated across a single layer of mucosal epithelial cells that form a critical barrier between the gut lumen and the underlying tissue. During the inflammatory response, neutrophils also contribute to the recruitment of other immune cells and facilitate mucosal healing by releasing mediators necessary for the resolution of inflammation. Although the above responses are clearly beneficial, excessive recruitment and accumulation of activated neutrophils in the intestine under pathological conditions such as inflammatory bowel disease is associated with mucosal injury and debilitating disease symptoms. Thus, depending on the circumstances, neutrophils can be viewed as either good or bad. In this article, we summarize the beneficial and deleterious roles of neutrophils in the intestine during health and disease and provide an overview of what is known about neutrophil function in the gut.


Subject(s)
Inflammation/immunology , Intestinal Diseases/immunology , Neutrophils/immunology , Animals , Humans , Inflammation/metabolism , Intestinal Diseases/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Neutrophil Activation/immunology , Neutrophils/metabolism
11.
Mucosal Immunol ; 2(1): 33-42, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19079332

ABSTRACT

Guanylate-binding protein-1 (GBP-1) is an interferon inducible large GTPase involved in endothelial cell proliferation and invasion. In this report, expression and function of GBP-1 were investigated in vitro in intestinal epithelia after exposure to interferon-gamma and in human colonic mucosa from individuals with inflammatory bowel disease (IBD). Interestingly, in contrast to other epithelia, GBP-1 distributed to the plasma membrane in intestinal epithelial cells where it colocalized with the tight junction protein coxsackie- and adenovirus receptor. In addition, expression of GBP-1 was upregulated in colonic epithelia of individuals with IBD. Downregulation of GBP-1 by siRNA resulted in enhanced permeability that correlated with increased apoptosis. Indeed, inhibition of caspase activity prevented the inhibition of barrier formation induced by the loss of GBP-1. These data suggest that GBP-1 is a novel marker of intestinal mucosal inflammation that may protect against epithelial apoptosis induced by inflammatory cytokines and subsequent loss of barrier function.


Subject(s)
Apoptosis/drug effects , Epithelial Cells/metabolism , GTP-Binding Proteins/metabolism , Interferon-gamma/pharmacology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Tight Junctions/metabolism , Apoptosis/immunology , Cell Line , Colon/immunology , Colon/metabolism , Down-Regulation/drug effects , Epithelial Cells/immunology , GTP-Binding Proteins/genetics , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , RNA, Small Interfering/genetics
13.
Oncogene ; 26(8): 1222-30, 2007 Feb 22.
Article in English | MEDLINE | ID: mdl-16924233

ABSTRACT

Although dysregulation of tight junction (TJ) proteins is observed in epithelial malignancy, their participation in epithelial transformation is poorly understood. Recently we demonstrated that expression of oncogenic Raf 1 in Pa4 epithelial cells disrupts TJs and induces an oncogenic phenotype by downregulating expression of the TJ protein, occludin. Here we report the mechanism by which Raf 1 regulates occludin expression. Raf 1 inhibited occludin transcription by repressing a minimal segment of the occludin promoter in concert with upregulation of the transcriptional repressor, Slug without influencing the well-documented transcriptional repressor, Snail. Overexpression of Slug in Pa4 cells recapitulated the effect of Raf 1 on occludin expression, and depletion of Slug by small interfering RNA abrogated the effect of Raf 1 on occludin. Finally, chromatin immunoprecipitation assays and site-directed mutagenesis demonstrated a direct interaction between Slug and an E-box within the minimal Raf 1-responsive segment of the occludin promoter. These findings support a role of Slug in mediating Raf 1-induced transcriptional repression of occludin and subsequent epithelial to mesenchymal transition.


Subject(s)
Membrane Proteins/genetics , Proto-Oncogene Proteins c-raf/metabolism , Repressor Proteins/metabolism , Tight Junctions/metabolism , Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Down-Regulation , E-Box Elements , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Humans , MAP Kinase Signaling System/drug effects , Membrane Proteins/analysis , Membrane Proteins/metabolism , Occludin , Rats , Snail Family Transcription Factors , Tight Junctions/chemistry , Zinc Fingers
14.
Am J Pathol ; 159(6): 2001-9, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733350

ABSTRACT

Inflammatory bowel disease (IBD) consisting of ulcerative colitis (UC) and Crohn's (CD) typically displays a waxing and waning course punctuated by disease flares that are characterized by transepithelial migration of neutrophils (PMN) and altered barrier function. Since epithelial barrier function is primarily regulated by the apical most intercellular junction referred to as the tight junction (TJ), our aim was to examine expression of TJ and adherens junction (AJ) proteins in relation to PMN infiltration in mucosal tissue samples from patients with active IBD. Expression of epithelial intercellular TJ proteins (occludin, ZO-1, claudin-1, and JAM) and subjacent AJ (beta-catenin and E-cadherin) proteins were examined by immunoflourescence/confocal microscopy, immunohistochemistry, and Western blotting. Colonic mucosa from patients with UC revealed dramatic, global down-regulation of the key TJ transmembrane protein occludin in regions of actively transmigrating PMN and in quiescent areas in the biopsy samples. Significant decreases in occludin expression were observed at the protein and mRNA levels by Western and Northern blotting. In contrast, expression of other TJ and AJ proteins such as ZO-1, claudin-1, JAM, beta-catenin, and E-cadherin were down-regulated only in epithelial cells immediately adjacent to transmigrating PMN. Analysis of inflamed mucosa from Crohn's disease patients mirrored the results obtained with UC patients. No change in TJ and AJ protein expression was observed in colonic epithelium from patients with collagenous colitis or lymphocytic colitis that are respectively characterized by a thickened subepithelial collagen plate and increased intraepithelial lymphocytes. These results suggest that occludin expression is diminished in IBD by mechanisms distinct from those regulating expression of other intercellular junction proteins. We speculate that down-regulation of epithelial occludin may play a role in enhanced paracellular permeability and PMN transmigration that is observed in active inflammatory bowel disease.


Subject(s)
Epithelium/pathology , Inflammatory Bowel Diseases/pathology , Intercellular Junctions/pathology , Neutrophils/pathology , Proteins/metabolism , Trans-Activators , Adherens Junctions/metabolism , Adherens Junctions/pathology , Cadherins/metabolism , Cell Adhesion Molecules/metabolism , Chronic Disease , Claudin-1 , Cytoskeletal Proteins/metabolism , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelium/metabolism , Gene Expression Regulation , Humans , Immunoblotting , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Intercellular Junctions/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Junctional Adhesion Molecules , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neutrophil Infiltration , Occludin , Phosphoproteins/metabolism , Proteins/genetics , RNA/genetics , RNA/metabolism , Tight Junctions/metabolism , Tight Junctions/pathology , Zonula Occludens-1 Protein , beta Catenin
15.
Am J Pathol ; 159(6): 2281-91, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733377

ABSTRACT

The combination of tumor necrosis factor (TNF)-alpha plus interferon (IFN)-gamma has been shown previously to promote redistribution of platelet/endothelial cell adhesion molecule-1 (PECAM-1) (CD31), junctional adhesion molecule (JAM), and VE-cadherin away from lateral junctions of human umbilical vein endothelial cell monolayers. In parallel, neutrophil transmigration was significantly reduced. Because PECAM-1 and JAM have been implicated in leukocyte transmigration, the observed redistribution by cytokine activation was presumed to represent the mechanism causing decreased transmigration under static conditions. The current results confirm that culture of human umbilical vein endothelial cells with TNF-alpha plus IFN-gamma caused a decrease in surface-expressed and junctional-localized JAM and PECAM-1, but did not cause decreased leukocyte transmigration in an in vitro flow assay. Furthermore, blocking monoclonal antibody to PECAM-1 still significantly reduced monocyte transmigration, demonstrating that it retains a functional role even though its levels were reduced and redistributed away from junctions, whereas a panel of monoclonal antibodies to JAM failed to reduce leukocyte transmigration. Given the alterations in junction protein location, permeability function was assessed. IFN-gamma alone or TNF-alpha plus IFN-gamma significantly increased permeability, but TNF-alpha alone did not, suggesting lack of correlation between transmigration and loss of permeability. In conclusion, cytokine activation induced loss and redistribution of PECAM-1 and JAM away from lateral junctions, but per se does not negatively regulate either neutrophil or monocyte transmigration under flow.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Endothelium, Vascular/drug effects , Interferon-gamma/pharmacology , Leukocytes/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Cell Adhesion/drug effects , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Flow Cytometry , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Leukocytes/cytology , Monocytes/cytology , Monocytes/drug effects , Pulsatile Flow
16.
Immunology ; 104(2): 185-97, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11683959

ABSTRACT

The expression of cell-surface antigens can delineate specific leucocyte developmental or functional stages. For example, certain membrane glycoproteins are expressed selectively on leucocyte subsets only after activation. Leucocyte activation can also induce changes in carbohydrate epitopes expressed on surface antigens. In the present studies, we report on a novel monoclonal immunoglobulin M antibody (mAb 13.22) that recognizes a unique carbohydrate epitope expressed on human leucocyte membrane proteins. Characterization of mAb 13.22 specificity by immunoblotting showed that it recognized proteins of MW approximately 95 000 and 150 000, including both CD18 and CD11b. The mAb 13.22 epitope was removed by N-glycosidase F but not by endoglycosidase H or fucosidase, demonstrating that it is an N-linked carbohydrate antigen. Interestingly, immunoblot staining was enhanced after neuraminidase treatment, suggesting that the antibody epitope might also be partially masked by sialic acid. In resting leucocytes, the mAb 13.22 antigen was expressed strongly on neutrophils, while dull staining was present on monocytes, and no lymphocyte staining was observed. In marked contrast, treatment of leucocytes with neuraminidase resulted in exposure of a mAb 13.22 neoepitope on a subset of lymphocytes (primarily T lymphocytes and natural killer cells) as well as up-regulated staining more than 18-fold on monocytes. Activation of lymphocytes in culture with phytohaemagglutinin or concanavalin A also unmasked the mAb 13.22 neoepitope on approximately 37% of the CD45RO+ lymphocytes. Furthermore, analysis of leucocytes collected from the synovial fluid of patients with rheumatoid arthritis showed that approximately 18% of the lymphocytes present expressed the mAb 13.22 neoepitope. Taken together, our results suggest that the mAb 13.22 carbohydrate neoepitope could represent a physiologically relevant marker that is up-regulated on leucocyte subsets during the inflammatory response.


Subject(s)
Epitopes/metabolism , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , Up-Regulation/immunology , Antibodies, Monoclonal/immunology , Blotting, Western , Carbohydrates/immunology , Cell Culture Techniques , Electrophoresis, Polyacrylamide Gel , Epitopes/drug effects , Humans , Lymphocyte Subsets/immunology , Neuraminidase/pharmacology , Synovial Fluid/immunology , Up-Regulation/drug effects
17.
Gastroenterology ; 121(3): 566-79, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11522741

ABSTRACT

BACKGROUND & AIMS: Tight junctions are crucial determinants of barrier function in polarized intestinal epithelia and are regulated by Rho guanosine triphosphatase. Rho kinase (ROCK) is a downstream effector of Rho. METHODS: A specific inhibitor of ROCK, Y-27632, was used to examine the role of ROCK in the regulation of tight junctions in model intestinal (T84) cells by electrophysiologic, biochemical, morphologic, and molecular biologic approaches. RESULTS: ROCK inhibition induced reorganization of apical F-actin structures and enhanced paracellular permeability but did not alter the distribution or detergent solubility of tight junction proteins. Confocal microscopy showed colocalization of a subpool of ROCK with the tight junction protein zonula occludens 1. Inhibition of ROCK function by a dominant negative mutant of ROCK also produced reorganization of apical F-actin structures without disruption of tight junctions. ROCK inhibition in calcium switch assays showed that ROCK is necessary for the assembly of tight and adherens junctions. Upon calcium repletion, occludin, zonula occludens 1, and E-cadherin failed to redistribute to the intercellular junctions; assembly of the apical F-actin cytoskeleton was prevented; and barrier function failed to recover. CONCLUSIONS: We suggest that ROCK regulates intact tight junctions via its effects on the F-actin cytoskeleton. ROCK is also critical for assembly of the apical junctional proteins and the F-actin cytoskeleton organization during junctional formation.


Subject(s)
Intestinal Mucosa/cytology , Intestinal Mucosa/enzymology , Protein Serine-Threonine Kinases/metabolism , Tight Junctions/enzymology , Actins/metabolism , Adherens Junctions/chemistry , Adherens Junctions/enzymology , Amides/pharmacology , Caco-2 Cells , Cadherins/analysis , Cell Membrane Permeability/physiology , Cell Polarity/physiology , Cells, Cultured , Detergents , Electrophysiology , Enzyme Inhibitors/pharmacology , Epithelial Cells/enzymology , Epithelial Cells/ultrastructure , Humans , Intestinal Absorption/physiology , Intracellular Signaling Peptides and Proteins , Membrane Proteins/analysis , Mutagenesis/physiology , Occludin , Octoxynol , Phosphoproteins/analysis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyridines/pharmacology , Solubility , Tight Junctions/chemistry , Transfection , Zonula Occludens-1 Protein , rho-Associated Kinases
18.
J Biol Chem ; 276(43): 40156-66, 2001 Oct 26.
Article in English | MEDLINE | ID: mdl-11479293

ABSTRACT

CD47, a cell surface glycoprotein, plays an important role in modulating neutrophil (PMN) migration across endothelial and epithelial monolayers. Here we show that anti-CD47 monoclonal antibodies (mAbs) delay PMN migration across collagen-coated filters or T84 epithelial monolayers toward the chemoattractant formylmethionylleucylphenylalanine (fMLP). Despite delayed transmigration by anti-CD47 mAbs, the numbers of PMN migrating across in either condition were the same as in the presence of control non-inhibitory mAbs. Cell surface labeling and immunoprecipitation demonstrated upregulation of CD47 to the PMN cell surface with kinetics similar to those of the transmigration response. Subcellular fractionation studies revealed redistribution of CD47 from intracellular compartments that co-sediment with secondary granules to plasma membrane-containing fractions after fMLP stimulation. Experiments performed to investigate potential signaling pathways revealed that inhibition of tyrosine phosphorylation with genistein reversed the anti-CD47-mediated PMN migration delay, whereas inhibition of phosphatidylinositol 3-kinase only partially reversed anti-CD47 effects that correlated with a rapid increase in PMN cell surface CD47. Analysis of the contribution of epithelial-expressed CD47 to PMN transmigration revealed that PMN migration across CD47-deficient epithelial monolayers (CaCO2) was significantly increased after stable transfection with CD47. These results suggest that cell surface CD47 and downstream tyrosine phosphorylation signaling events regulate, in part, the rate of PMN migration during the inflammatory response.


Subject(s)
Antigens, CD/metabolism , Carrier Proteins/metabolism , Chemotaxis, Leukocyte/physiology , Neutrophils/physiology , Antibodies, Monoclonal/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , CD47 Antigen , Caco-2 Cells , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Compartmentation , Cell Fractionation , Chemotaxis, Leukocyte/drug effects , Diffusion Chambers, Culture , Genistein/pharmacology , Humans , Inflammation , Intestinal Mucosa/cytology , Isoflavones/pharmacology , Models, Biological , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Recombinant Proteins/metabolism , Signal Transduction , Up-Regulation
19.
J Immunol ; 166(12): 7250-9, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11390474

ABSTRACT

Recently, we identified a neutrophil-binding phage displaying a novel peptide motif, GPNLTGRW. It was determined that this peptide, when displayed on bacteriophage (FGP phage), elicits a transient increase in cytosolic calcium. Here, we show that FGP phage stimulate neutrophil chemotaxis and induce a pertussis toxin-sensitive rise in cytosolic calcium in monocytes as well as in neutrophils. In contrast to the calcium response elicited by classical chemoattractants fMLP and IL-8, the FGP phage-elicited response in neutrophils is dependent on extracellular calcium and is mediated by receptor-activated, divalent cation channels. Consistent with G protein-coupled receptor signaling, FGP phage effect homologous and reciprocal heterologous desensitization with fMLP- and IL-8-stimulated calcium responses. Like non-G protein-coupled responses, the FGP-elicited calcium transient is abolished with phosphoinositide-3-kinase inactivation. Nonetheless, specific binding of GTP to neutrophil membranes follows stimulation with FGP phage, further supporting involvement of G proteins. However, FGP phage neither bind to nor elicit a calcium response from transfectant cells harboring known candidate G protein-coupled receptors. These data together suggest that the elicited responses are mediated by a novel G protein-coupled receptor or represent novel responses of a known receptor.


Subject(s)
Bacteriophages/immunology , Chemotaxis, Leukocyte , GTP-Binding Proteins/metabolism , Oligopeptides/immunology , Oligopeptides/metabolism , Animals , CHO Cells , Calcium Channels/metabolism , Calcium Signaling/genetics , Calcium Signaling/immunology , Cations, Divalent/metabolism , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Chemotaxis, Leukocyte/genetics , Cricetinae , Extracellular Space/physiology , GTP-Binding Proteins/genetics , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Interleukin-8/physiology , Lymphocytes/metabolism , Mice , Monocytes/immunology , Monocytes/metabolism , N-Formylmethionine Leucyl-Phenylalanine/metabolism , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/metabolism , Oligopeptides/genetics , Peptide Library , Pertussis Toxin , Phosphatidylinositol 3-Kinases/physiology , Protein Binding/genetics , Protein Binding/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/physiology , Transfection , Virulence Factors, Bordetella/pharmacology
20.
Am J Physiol Gastrointest Liver Physiol ; 280(4): G746-54, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11254502

ABSTRACT

Neutrophil (PMN) transepithelial migration is a major effector of epithelial defense in inflammatory diseases involving mucosal surfaces. However, major receptor-ligand interactions between epithelial cells and PMN remain incompletely characterized. To better define the molecular events involved in PMN interactions with epithelial cells, we produced a monoclonal antibody called g82 that inhibited PMN transepithelial migration in the physiological basolateral-to-apical direction. The g82 antigen localized to the apical surface of human colonic epithelium and was significantly upregulated under inflammatory conditions. Immunoprecipitation revealed two polypeptides of M(r) 207 and 32 kDa. F(ab')(2) fragments from g82 IgG had no effect on transmigration, suggesting Fc dependence. Further experiments confirmed dependence on the PMN Fc receptor CD32A and that the observed effects were secondary to a failure of PMN to detach from the apical epithelial surface. These Fc-mediated events were epitope specific since binding, isotype-matched antibodies did not affect detachment. These results identify a new mechanism for retention of PMN at the apical epithelial surface following transepithelial migration. This pathway may be important in pathogen clearance and mucosal pathophysiology associated with autoimmunity.


Subject(s)
Immunoglobulin Fc Fragments/physiology , Neutrophil Infiltration/physiology , Neutrophils/physiology , Animals , Buffers , Cell Adhesion/physiology , Cells, Cultured , Colon/cytology , Colon/immunology , Epithelial Cells/physiology , Epithelium/physiology , Fluorescent Antibody Technique, Direct , Humans , Immunoglobulin Fc Fragments/biosynthesis , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Precipitin Tests
SELECTION OF CITATIONS
SEARCH DETAIL
...