Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mucosal Immunol ; 7(6): 1312-25, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24670424

ABSTRACT

It is well established that polymorphisms of the caspase activation and recruitment domain 15 (CARD15) gene, a major risk factor in Crohn's disease (CD), lead to loss of nucleotide-binding oligomerization domain 2 (NOD2) function. However, a molecular explanation of how such loss of function leads to increased susceptibility to CD has remained unclear. In a previous study exploring this question, we reported that activation of NOD2 in human dendritic cells by its ligand, muramyl dipeptide (MDP), negatively regulates Toll-like receptor (TLR)-mediated inflammatory responses. Here we show that NOD2 activation results in increased interferon regulatory factor 4 (IRF4) expression and binding to tumor necrosis factor receptor associated factor 6 (TRAF6) and RICK (receptor interacting serine-threonine kinase). We then show that such binding leads to IRF4-mediated inhibition of Lys63-linked polyubiquitination of TRAF6 and RICK and thus to downregulation of nuclear factor (NF)-κB activation. Finally, we demonstrate that protection of mice from the development of experimental colitis by MDP or IRF4 administration is accompanied by similar IRF4-mediated effects on polyubiquitination of TRAF6 and RICK in colonic lamina propria mononuclear cells. These findings thus define a mechanism of NOD2-mediated regulation of innate immune responses to intestinal microflora that could explain the relation of CARD15 polymorphisms and resultant NOD2 dysfunction to CD.


Subject(s)
Colon/immunology , Crohn Disease/immunology , Down-Regulation/immunology , Interferon Regulatory Factors/immunology , Nod2 Signaling Adaptor Protein/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , TNF Receptor-Associated Factor 6/immunology , Ubiquitination/immunology , Animals , Colon/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Dendritic Cells/immunology , Dendritic Cells/pathology , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interferon Regulatory Factors/genetics , Mice , Nod2 Signaling Adaptor Protein/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , TNF Receptor-Associated Factor 6/genetics , Ubiquitination/genetics
2.
Mucosal Immunol ; 7(2): 348-58, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23945545

ABSTRACT

Transforming growth factor-ßs (TGF-ßs) are secreted from cells as latent complexes and the activity of TGF-ßs is controlled predominantly through activation of these complexes. Tolerance to the fetal allograft is essential for pregnancy success; TGF-ß1 and TGF-ß2 play important roles in regulating these processes. Pregnancy-specific ß-glycoproteins (PSGs) are present in the maternal circulation at a high concentration throughout pregnancy and have been proposed to have anti-inflammatory functions. We found that recombinant and native PSG1 activate TGF-ß1 and TGF-ß2 in vitro. Consistent with these findings, administration of PSG1 protected mice from dextran sodium sulfate (DSS)-induced colitis, reduced the secretion of pro-inflammatory cytokines, and increased the number of T regulatory cells. The PSG1-mediated protection was greatly inhibited by the coadministration of neutralizing anti-TGF-ß antibody. Our results indicate that proteins secreted by the placenta directly contribute to the generation of active TGF-ß and identify PSG1 as one of the few known biological activators of TGF-ß2.


Subject(s)
Colitis/metabolism , Colitis/prevention & control , Pregnancy-Specific beta 1-Glycoproteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colitis/chemically induced , Colitis/immunology , Cytokines/biosynthesis , Dextran Sulfate/adverse effects , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice , Pregnancy-Specific beta 1-Glycoproteins/administration & dosage , Protein Binding , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta2/metabolism
3.
Mucosal Immunol ; 7(3): 521-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24064669

ABSTRACT

In the present study we investigated the molecular mechanisms regulating the expression of RAR-related orphan receptor gamma t (RORγt), the central factor controlling interleukin (IL)-17 transcription and Th17 differentiation. In key studies, we found that cells from mice with major deletions of E-protein transcription factors, E2A and HEB, display greatly reduced RORγt/IL-17 expression and that E-protein-deficient mice exhibit greatly diminished IL-17-dependent inflammation in experimental allergic encephalitis models. In additional studies, we unexpectedly found that cells from mice with deletion of Id3, a protein that inhibits E-protein binding to DNA, display diminished RORγt/IL-17 expression and mice deficient in this protein exhibit decreased Th17-mediated inflammation in a cell-transfer colitis model. The explanation of these initially paradoxical findings came from studies showing that Id3 deficiency leads to increased IL-4-induced GATA-3 expression, the latter a negative regulator of RORγt transcription; thus, increased Id3 expression likely has a net positive effect on RORγt expression via its inhibition of IL-4 production. Finally, we found that both E-proteins and Id3 are upregulated in tandem by the cytokines that induce Th17 differentiation, transforming growth factor-ß, and IL-6, implying that these transcription factors are critical regulators of Th17 induction.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Th17 Cells/immunology , Th17 Cells/metabolism , Transcription, Genetic , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Gene Expression , Genes, Reporter , Inhibitor of Differentiation Protein 1/metabolism , Interleukin-17/genetics , Interleukin-17/metabolism , Interleukin-6/metabolism , Mice , Mice, Knockout , Promoter Regions, Genetic , Signal Transduction , Th17 Cells/cytology , Transforming Growth Factor beta/metabolism
4.
Mucosal Immunol ; 4(2): 172-85, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20980995

ABSTRACT

The tumor necrosis factor (TNF)-family cytokine TL1A (TNFSF15) costimulates T cells through its receptor DR3 (TNFRSF25) and is required for autoimmune pathology driven by diverse T-cell subsets. TL1A has been linked to human inflammatory bowel disease (IBD), but its pathogenic role is not known. We generated transgenic mice that constitutively express TL1A in T cells or dendritic cells. These mice spontaneously develop IL-13-dependent inflammatory small bowel pathology that strikingly resembles the intestinal response to nematode infections. These changes were dependent on the presence of a polyclonal T-cell receptor (TCR) repertoire, suggesting that they are driven by components in the intestinal flora. Forkhead box P3 (FoxP3)-positive regulatory T cells (Tregs) were present in increased numbers despite the fact that TL1A suppresses the generation of inducible Tregs. Finally, blocking TL1A-DR3 interactions abrogates 2,4,6 trinitrobenzenesulfonic acid (TNBS) colitis, indicating that these interactions influence other causes of intestinal inflammation as well. These results establish a novel link between TL1A and interleukin 13 (IL-13) responses that results in small intestinal inflammation, and also establish that TL1A-DR3 interactions are necessary and sufficient for T cell-dependent IBD.


Subject(s)
Enteritis/immunology , Interleukin-13/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Animals , CD2 Antigens/genetics , CD2 Antigens/immunology , Colitis/immunology , Colitis/pathology , Dendritic Cells/immunology , Dose-Response Relationship, Immunologic , Enteritis/pathology , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/immunology , Gene Order , HEK293 Cells , Humans , Immunologic Memory/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin-13/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Receptors, Tumor Necrosis Factor, Member 25/metabolism , T-Lymphocytes
5.
Anaesthesist ; 59(6): 519-23, 2010 Jun.
Article in German | MEDLINE | ID: mdl-20458453

ABSTRACT

BACKGROUND: Venepuncture is one of the most stressful situations for children during induction of general anesthesia. Therefore, many clinicians use a local anesthesia patch (EMLA) containing a mixture of lidocaine and prilocaine in order to reduce the stress for pediatric patients. This study compared the effect of a new heated topical anesthesia delivery system containing lidocaine and tetracaine (Rapydan) with the lidocaine/prilocaine patch EMLA. METHODS: The study design was prospective, randomized, single-blinded and monocenter. A total of 200 children aged from 3 to 13 years were randomized into group E (EMLA) or group R (Rapydan). The primary endpoint of the study was the overall incidence of pain. Additionally, the intensity of pain during venous puncture was evaluated by means of an investigator-based 4 point pain score: 0 no reaction, 1 gentle movement/grimacing, 2 moderate withdrawal of the arm/crying and 3 strong withdrawal/screaming. Furthermore, erythema of the skin, visibility of the veins and success rate of the punctures were assessed. RESULTS: Mean contact time of the patch with the skin was 35 min in both groups. The overall incidence of pain was 46% in group E and 12% in group R (p<0.001). The intensity of pain also differed significantly between the groups. A pain score of 1 was observed in 24% (group E) versus 10% (group R), a score of 2 was documented in 13% (group E) versus 1% (group R) and a score of 3 was observed in 9% (group E) versus 1% (group R; p<0.001). Erythema of the skin was observed more frequently in group R (p<0.001). Visibility of the veins and success rate of venous puncture did not differ significantly. CONCLUSIONS: After a contact time of 35 min the Rapydan patch led to superior analgesia during venous puncture than the EMLA patch. With regard to visibility of the veins and success rate of the punctures, differences between the two patches were not observed.


Subject(s)
Anesthesia, Local , Anesthetics, Local , Lidocaine , Phlebotomy/adverse effects , Prilocaine , Tetracaine , Administration, Cutaneous , Adolescent , Anesthetics, Local/administration & dosage , Behavior , Child , Child, Preschool , Double-Blind Method , Endpoint Determination , Female , Humans , Lidocaine/administration & dosage , Lidocaine, Prilocaine Drug Combination , Male , Pain/prevention & control , Pain Measurement/drug effects , Prilocaine/administration & dosage , Prospective Studies , Tetracaine/administration & dosage
6.
Clin Exp Immunol ; 155(3): 447-56, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19094118

ABSTRACT

Granulocyte-colony stimulating factor (G-CSF) has proved to be a successful therapy for some patients with Crohn's disease. Given the known ability of G-CSF to exert anti-T helper 1 effects and to induce interleukin (IL)-10-secreting regulatory T cells, we studied whether clinical benefit from G-CSF therapy in active Crohn's disease was associated with decreased inflammatory cytokine production and/or increased regulatory responses. Crohn's patients were treated with G-CSF (5 microg/kg/day subcutaneously) for 4 weeks and changes in cell phenotype, cytokine production and dendritic cell subsets were measured in the peripheral blood and colonic mucosal biopsies using flow cytometry, enzyme-linked immunosorbent assay and immunocytochemistry. Crohn's patients who achieved a clinical response or remission based on the decrease in the Crohn's disease activity index differed from non-responding patients in several important ways: at the end of treatment, responding patients had significantly more CD4(+) memory T cells producing IL-10 in the peripheral blood; they also had a greatly enhanced CD123(+) plasmacytoid dendritic cell infiltration of the lamina propria. Interferon-gamma production capacity was not changed significantly except in non-responders, where it increased. These data show that clinical benefit from G-CSF treatment in Crohn's disease is accompanied by significant induction of IL-10 secreting T cells as well as increases in plasmacytoid dendritic cells in the lamina propria of the inflamed gut mucosa.


Subject(s)
Crohn Disease/drug therapy , Dendritic Cells/immunology , Granulocyte Colony-Stimulating Factor/therapeutic use , Interleukin-10/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Crohn Disease/immunology , Cytokines/immunology , Drug Administration Schedule , Female , Humans , Immunohistochemistry , Immunophenotyping , Lymphocyte Activation/immunology , Male , Mucous Membrane/immunology , Pilot Projects , Recombinant Proteins , Statistics, Nonparametric , Treatment Outcome , Young Adult
7.
Mucosal Immunol ; 1 Suppl 1: S31-3, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19079225

ABSTRACT

Recent studies that have evaluated the immunologic factors that mediate the development of the two forms of inflammatory bowel disease, namely Crohn's disease and ulcerative colitis (UC), have suggested that these diseases are because of disparate immune responses. Although Crohn's disease has been characterized as a dysregulation of the T helper (Th)1/Th17 pathways more recent evidence has emerged that UC pathogenesis is associated with a nonclassical NK (natural killer) T cell producing an atypical Th2 (interleukin (IL)-13) response. In the following review the insights gained from both animal models and human studies as to the function that IL-13 and NK T cells have in the pathogenesis of UC will be discussed.


Subject(s)
Colitis, Ulcerative/immunology , Interleukin-13/immunology , Natural Killer T-Cells/immunology , Animals , Colitis, Ulcerative/chemically induced , Humans , Oxazolone/pharmacology
8.
Mucosal Immunol ; 1 Suppl 1: S5-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19079230

ABSTRACT

Nucleotide oligomerization domain (NOD)2 is a member of the NOD-like receptor family of proteins that initiate inflammatory responses when exposed to ligands derived from bacterial components that gain access to the intracellular milieu. It is thus somewhat paradoxical that polymorphisms in the gene that encode NOD2 (CARD15) that lead to impaired NOD2 function, are susceptibility factors in Crohn's disease, a condition marked by excessive inflammatory responses to normal bacterial flora. In an initial series of studies conducted in our laboratory to better define NOD2 function and to resolve this paradox we showed that NOD2 activation by its ligand, muramyl dipeptide (MDP) ordinarily downregulates responses to Toll-like receptor (TLR) stimulation, and thus cells lacking NOD2 mount increased responses to such stimulation. This fits with the fact that mice bearing an NOD2 transgene, and thus having cells with increased NOD2 function display decreased responses to TLR stimulation and are resistant to experimental colitis induction. In further studies, we showed that prestimulation of cells with NOD2 ligand renders them unresponsive to TLR stimulation, because such prestimulation results in the elaboration of inhibitory factor (IRF4), an inhibitor of TLR-induced inflammatory pathways. Furthermore, administration of MDP to normal mice induces IRF4 and prevents experimental colitis. These studies strongly suggest that NOD2 polymorphisms are associated with Crohn's disease because they lead to a decrease in the negative regulation of TLR responses occurring in the normal gut, and thus a pathologic increase in responses to the normal flora. The finding that MDP administration prevents experimental colitis opens the door to the possibility that such treatment might quell Crohn's disease relapses in patients without NOD2 abnormalities.


Subject(s)
Crohn Disease/genetics , Crohn Disease/metabolism , Genetic Predisposition to Disease/genetics , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Animals , Gene Expression Regulation , Humans , Mutation/genetics , Nod2 Signaling Adaptor Protein/deficiency , Toll-Like Receptors/metabolism
9.
Acta Odontol Scand ; 59(4): 244-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11570528

ABSTRACT

Studies conducted over the past 10 years have provided ample evidence that many types of inflammations arising from basic abnormalities of immune regulation are ultimately 'funneled' through a Th1 or Th2 T cell-mediated immune reaction. Thus, by understanding these types of reactions and, in particular, by identifying their natural checkpoints, one can control the inflammation regardless of its more basic causes. A case in point is the inflammatory disease of the intestine known as Crohn disease, a disease now thought to be due to one or more abnormalities leading to an excessive immune response to elements of the bacterial microflora of the gut. Both in murine models and by study of Crohn disease itself, we have shown that Crohn inflammation is due to a Th1 T-cell abnormality involving overproduction of interleukin (IL)-12, interferon (IFN-gamma, and tumor necrosis factor (TNF)-alpha. In addition, we and others have shown that treatment of mice with anti-IL-12 or other agents that downregulate the level of IL- 12 secretion can have a dramatic effect on the inflammation. This is because anti-IL-12 administration leads to apoptosis of activated Th1 T cells. A second checkpoint of Th1 T-cell-mediated inflammation involves its downregulation by the suppressor cytokine, transforming growth factor (TGF)-beta. We have been delivering TGF-beta to mice with experimental intestinal inflammation, using several novel approaches. In particular, we have successfully treated such mice with intranasally administered DNA encoding active TGF-beta. Another approach currently under investigation is delivery of TGF-beta by gene therapy. These and other developments in the understanding of inflammation paint a bright future for cytokine-based therapeutic agents. It is now apparent that these therapies are not only effective and safe but also potentially long-lasting.


Subject(s)
Colitis/immunology , Colitis/therapy , Immunotherapy/methods , Animals , Colitis/chemically induced , Down-Regulation , Gene Transfer Techniques , Interleukin-10/therapeutic use , Interleukin-12/antagonists & inhibitors , Intestinal Mucosa/immunology , Mice , Mice, Inbred Strains , T-Lymphocyte Subsets/immunology , Th1 Cells/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/therapeutic use , Trinitrobenzenesulfonic Acid , Tumor Necrosis Factor-alpha/antagonists & inhibitors
10.
Blood ; 97(10): 3161-70, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342444

ABSTRACT

Autoimmune lymphoproliferative syndrome (ALPS) is an inherited disorder in which genetic defects in proteins that mediate lymphocyte apoptosis, most often Fas, are associated with enlargement of lymph nodes and the spleen and a variety of autoimmune manifestations. Some patients with ALPS have relatives with these same apoptotic defects, however, who are clinically well. This study showed that the circulating levels of interleukin 10 (IL-10) were significantly higher (P <.001) in 21 patients with ALPS than in healthy controls. Moreover, the peripheral blood mononuclear cells (PBMCs) and lymphoid tissues of these patients with ALPS contained significantly higher levels of IL-10 messenger RNA (mRNA; P <.001 and P <.01, respectively). By fractionating PBMC populations, disproportionately high concentrations of IL-10 mRNA were found in the CD4(-)CD8(-) T-cell population, expansion of which is virtually pathognomonic for ALPS. Immunohistochemical staining showed intense IL-10 protein signals in lymph node regions known to contain CD4(-)CD8(-) T cells. Nonetheless, in vitro studies showed no influence of IL-10 on the survival of CD4(-)CD8(-) T cells. Overexpression of IL-10 in patients with inherited apoptotic defects is strongly associated with the overt manifestations of ALPS.


Subject(s)
Autoimmune Diseases/metabolism , Interleukin-10/metabolism , Lymphoid Tissue/metabolism , Lymphoproliferative Disorders/immunology , Apoptosis , Autoimmune Diseases/blood , Autoimmune Diseases/genetics , CD4 Antigens/analysis , CD8 Antigens/analysis , Cell Survival , Child , Child, Preschool , Female , Humans , Infant , Interleukin-10/blood , Interleukin-10/genetics , Leukocytes, Mononuclear/chemistry , Lymphoproliferative Disorders/blood , Lymphoproliferative Disorders/genetics , Male , RNA, Messenger/blood , T-Lymphocytes/immunology , T-Lymphocytes/physiology
11.
Trends Immunol ; 22(1): 21-6, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11286687

ABSTRACT

T-cell resistance against apoptosis contributes to inappropriate T-cell accumulation and the perpetuation of chronic mucosal inflammation in inflammatory bowel diseases (IBDs). Anti-interleukin-12 (IL-12) and anti-IL-6 receptor antibodies suppress colitis activity by the induction of T-cell apoptosis. These findings have important implications for the design of effective treatment regimens in IBD.


Subject(s)
Apoptosis/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Animals , Cell Death/immunology , Humans , Immunity, Mucosal
12.
J Immunol ; 166(5): 3522-32, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11207312

ABSTRACT

Trinitrobenzene sulfonic acid (TNBS)-induced colitis is an IL-12-driven, Th1 T cell-mediated colitis that resembles human Crohn's disease. In the present study, we showed initially that the oral administration of recombinant subunit B of cholera toxin (rCT-B) at the time of TNBS-induced colitis by intrarectal TNBS instillation inhibits the development of colitis or, at later time when TNBS-induced colitis is well established, brings about resolution of the colitis. Dose-response studies showed that a majority of mice (68%) treated with rCT-B at a dose of 100 microg (times four daily doses) exhibited complete inhibition of the development of colitis, whereas a minority (30%) treated with rCT-B at a dose of 10 microg (times four daily doses) exhibited complete inhibition; in both cases, however, the remaining mice exhibited some reduction in the severity of inflammation. In further studies, we showed that rCT-B administration is accompanied by prevention/reversal of increased IFN-gamma secretion (the hallmark of a Th1 response) without at the same time causing an increase in IL-4 secretion. This decreased IFN-gamma secretion was not associated with the up-regulation of the secretion of counterregulatory cytokines (IL-10 or TGF-beta), but was associated with a marked inhibition of IL-12 secretion, i.e., the secretion of the cytokine driving the Th1 response. Finally, we showed that rCT-B administration results in increased apoptosis of lamina propria cells, an effect previously shown to be indicative of IL-12 deprivation. From these studies, rCT-B emerges as a powerful inhibitor of Th1 T cell-driven inflammation that can conceivably be applied to the treatment of Crohn's disease.


Subject(s)
Cholera Toxin/administration & dosage , Cholera Toxin/immunology , Colitis/immunology , Colitis/prevention & control , Interleukin-12/antagonists & inhibitors , Interleukin-12/physiology , Trinitrobenzenesulfonic Acid , Administration, Oral , Administration, Rectal , Animals , Apoptosis/immunology , Cells, Cultured , Colitis/chemically induced , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , Disease Progression , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Interleukin-4/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred Strains , Oxazolone/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Transforming Growth Factor beta/biosynthesis , Trinitrobenzenesulfonic Acid/administration & dosage , Trinitrobenzenesulfonic Acid/antagonists & inhibitors
13.
Clin Immunol ; 97(2): 121-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11027452

ABSTRACT

Two patients with Bruton's X-linked agammaglobulinemia are described with bacteremia and skin/bone infection due to an organism which by 16S rRNA gene sequence analysis was most closely related to "Flexispira" rappini (and thus designated a Flexispira-like organism, FLO) and more distantly related to the Helicobacter species. The organism required microaerobic conditions and, supplemental H(2) gas for growth and was reliably stained with acridine orange. In common with Helicobacter cinaedi infections, the focus of the FLO infection was in one case in the blood vessels or lymphatics of an extremity and in the other case in the skin and adjacent bone of an extremity. In both cases, prolonged IV antibiotic therapy was necessary to clear the infection. The susceptibility of XLA patients to FLO infection appears to be related to the fact that XLA is associated with severe B cell (humoral) immunodeficiency and thus these patients have difficulty with intravascular or intralymphatic infection. These findings elucidate the nature of FLO infections in humans and point the way to their detection and treatment.


Subject(s)
Agammaglobulinemia/genetics , Bacteremia/complications , Bone Diseases/complications , Genetic Linkage , Helicobacter Infections/complications , Skin Diseases, Bacterial/complications , X Chromosome , Adult , Agammaglobulinemia/microbiology , Bone Diseases/immunology , Genes, rRNA/genetics , Helicobacter/genetics , Helicobacter Infections/immunology , Humans , Male , Sequence Analysis, RNA , Skin Diseases, Bacterial/immunology
14.
J Exp Med ; 192(1): 41-52, 2000 Jul 03.
Article in English | MEDLINE | ID: mdl-10880525

ABSTRACT

In this study, we show that a single intranasal dose of a plasmid encoding active transforming growth factor beta1 (pCMV-TGF-beta1) prevents the development of T helper cell type 1 (Th1)-mediated experimental colitis induced by the haptenating reagent, 2,4, 6-trinitrobenzene sulfonic acid (TNBS). In addition, such plasmid administration abrogates TNBS colitis after it has been established, whereas, in contrast, intraperitoneal administration of rTGF-beta1 protein does not have this effect. Intranasal pCMV-TGF-beta1 administration leads to the expression of TGF-beta1 mRNA in the intestinal lamina propria and spleen for 2 wk, as well as the appearance of TGF-beta1-producing T cells and macrophages in these tissues, and is not associated with the appearances of fibrosis. These cells cause marked suppression of interleukin (IL)-12 and interferon (IFN)-gamma production and enhancement of IL-10 production; in addition, they inhibit IL-12 receptor beta2 (IL-12Rbeta2) chain expression. Coadministration of anti-IL-10 at the time of pCMV-TGF-beta1 administration prevents the enhancement of IL-10 production and reverses the suppression of IL-12 but not IFN-gamma secretion. However, anti-IL-10 leads to increased tumor necrosis factor alpha production, especially in established colitis. Taken together, these studies show that TGF-beta1 inhibition of a Th1-mediated colitis is due to: (a) suppression of IL-12 secretion by IL-10 induction and (b) inhibition of IL-12 signaling via downregulation of IL-12Rbeta2 chain expression. In addition, TGF-beta1 may also have an inhibitory effect on IFN-gamma transcription.


Subject(s)
Colitis/therapy , Genetic Therapy , Plasmids/administration & dosage , Th1 Cells/immunology , Transforming Growth Factor beta/genetics , Administration, Intranasal , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/prevention & control , Colon/pathology , Cytokines/biosynthesis , Cytomegalovirus , Injections, Intraperitoneal , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred Strains , Recombinant Proteins/therapeutic use , Swine , Th1 Cells/drug effects , Transforming Growth Factor beta/therapeutic use , Trinitrobenzenesulfonic Acid
15.
Int Rev Immunol ; 19(1): 51-62, 2000.
Article in English | MEDLINE | ID: mdl-10723677

ABSTRACT

Disease states, such as the occurrence of gastrointestinal inflammation (Crohn's disease and ulcerative colitis), can be secondary to a host of determinants that act in conjunction to bring about pathologic change. The underlying factors that mediate the development of such mucosal inflammation has recently been brought to the forefront with the advent of animal models. The examination of these animal models have given researchers a better understanding of the mechanisms involved in the pathogenesis of inflammatory bowel disease. This review discusses one such model, TNBS-colitis, and the insights that it provides into the occurrence of IBD and its future treatment.


Subject(s)
Colitis , Disease Models, Animal , Trinitrobenzenesulfonic Acid , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/immunology , Colitis/pathology , Crohn Disease/immunology , Humans
16.
Gastroenterology ; 117(5): 1078-88, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10535870

ABSTRACT

BACKGROUND & AIMS: Trinitrobenzene sulfonic acid (TNBS)-induced colitis is a T-helper 1 (Th1) T cell-mediated inflammation that is rapidly reversed by administration of anti-interleukin (IL) 12. This study sought to define the mechanism of this curative effect. METHODS: Cells and tissue from mice with TNBS colitis receiving treatment with anticytokines were analyzed for phenotype, cytokine production, and apoptosis. RESULTS: In initial studies, we found that treatment of mice with TNBS-induced colitis with anti-IL-12 was more effective than with anti-interferon (IFN)-gamma, and that anti-IL-12 led to complete normalization of IFN-gamma production by lamina propria T cells ex vivo, whereas anti-IFN-gamma did not. These data suggesting that anti-IL-12 leads to reversal of colitis by elimination of the Th1 T cells were substantiated by studies showing that anti-IL-12 treatment led to increased numbers of apoptotic cells in the lamina propria and spleen by both TUNEL staining of tissues and dispersed spleen cell populations. Finally, we found that the observed apoptosis was mediated by the Fas pathway because (1) MRL/MpJ-lpr(fas) mice lacking Fas function develop colitis that responds poorly to treatment with anti-IL-12; and (2) SJL/J mice with TNBS colitis that received Fas-Fc to block the Fas pathway were resistant to anti-IL-12 treatment. CONCLUSIONS: These studies show that a main mechanism of action of anti-IL-12 in TNBS-induced colitis is the induction of Fas-mediated apoptosis of the Th1 T cells, causing inflammation.


Subject(s)
Antibodies/therapeutic use , Apoptosis/drug effects , Colitis/drug therapy , Interleukin-12/immunology , Th1 Cells/drug effects , Animals , Colitis/chemically induced , Colitis/physiopathology , Interferon-gamma/immunology , Male , Mice , Mice, Inbred MRL lpr , Mice, Inbred Strains , T-Lymphocytes/drug effects , T-Lymphocytes/physiology , Th1 Cells/physiology , Trinitrobenzenesulfonic Acid , fas Receptor/metabolism , fas Receptor/physiology
18.
Eur J Immunol ; 28(12): 4275-85, 1998 12.
Article in English | MEDLINE | ID: mdl-9862365

ABSTRACT

Intestinal lymphangiectasia (InL) is a disease characterized by hypoproteinemia and lymphocytopenia resulting from blocked intestinal lymphatics and loss of lymph fluid into the gastrointestinal tract. This leads to immunologic abnormalities including hypogammaglobulinemia, skin anergy and impaired allograft rejection. In the present study, we evaluated whether the above immunologic abnormalities are secondary to a quantitative or qualitative disorder of T cells. In initial studies we demonstrated that adult InL patients' peripheral blood contain strikingly (and significantly) reduced numbers of CD4+/CD45RA+ T cells, whereas the numbers of CD4+/CD45RO+ T cells were only moderately (and not significantly) reduced. In addition, the CD4+/CD45RO+ T cell population contained an increased percentage of highly differentiated and previously sensitized cells, as demonstrated by decreased CD27 and CD31 expression and increased HLA-DR and CD69 expression. In subsequent functional studies, we showed that the InL CD4+/CD45RO+ T cells, when stimulated in vitro, proliferate fivefold less than control CD4+/CD45RO+ T cells and produce fourfold more IL-4 and threefold less IFN-gamma and IL-2. Thus, this cytokine production profile also reflects the highly differentiated nature of the residual cell population. Overall, these studies provide new information on the trafficking of naive/mature and Th1/Th2 T cell populations in this disease model.


Subject(s)
Intestines/immunology , Leukocyte Common Antigens/immunology , Lymphangiectasis, Intestinal/immunology , Adult , Humans , Immunity, Mucosal/immunology , Immunophenotyping , Intestines/pathology , Lymphangiectasis, Intestinal/pathology , Lymphocytes/immunology , Lymphocytes/pathology , Middle Aged
20.
J Exp Med ; 188(10): 1929-39, 1998 Nov 16.
Article in English | MEDLINE | ID: mdl-9815270

ABSTRACT

In this study we describe oxazolone colitis, a new form of experimental colitis. This model is induced in SJL/J mice by the rectal instillation of the haptenating agent, oxazolone, and is characterized by a rapidly developing colitis confined to the distal half of the colon; it consists of a mixed neutrophil/lymphocyte infiltration limited to the superficial layer of the mucosa which is associated with ulceration. Oxazolone colitis is a T helper cell type 2 (Th2)-mediated process since stimulated T cells from lesional tissue produce markedly increased amounts of interleukin (IL)-4 and IL-5; in addition, anti-IL-4 administration leads to a striking amelioration of disease, whereas anti-IL-12 administration either has no effect or exacerbates disease. Finally, this proinflammatory Th2 cytokine response is counterbalanced by a massive transforming growth factor-beta (TGF-beta) response which limits both the extent and duration of disease: lesional (distal) T cells manifest a 20-30-fold increase in TGF-beta production, whereas nonlesional (proximal) T cells manifest an even greater 40-50-fold increase. In addition, anti-TGF-beta administration leads to more severe inflammation which now involves the entire colon. The histologic features and distribution of oxazolone colitis have characteristics that resemble ulcerative colitis (UC) and thus sharply distinguish this model from most other models, which usually resemble Crohn's disease. This feature of oxazolone colitis as well as its cytokine profile have important implications to the pathogenesis and treatment of UC.


Subject(s)
Antibodies/therapeutic use , Colitis/chemically induced , Interleukin-4/immunology , Oxazolone/pharmacology , Th2 Cells/immunology , Administration, Rectal , Animals , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/pathology , Cytokines/metabolism , Disease Models, Animal , Histocytochemistry , Humans , Inflammation/immunology , Interleukins/metabolism , Mice , Mice, Inbred Strains , Oxazolone/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...