Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Gastroenterology ; 140(2): 550-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21056041

ABSTRACT

BACKGROUND & AIMS: Interactions between lymphocytes and intestinal epithelial cells occur in the subepithelial space of the gastrointestinal tract. Normal human lamina propria lymphocytes (LPLs) induce differentiation of intestinal epithelial cells. The absence of LPLs in mice, such as in RAG1(-/-) mice, results in defects in epithelial cell differentiation. We investigated the role of lymphoepithelial interactions in epithelial differentiation and barrier function. METHODS: We used adoptive transfer to determine if CD4(+) T cells (CD4(+)CD62L(+)CD45Rb(Hi) and/or CD4(+)CD62L(+)CD45Rb(Lo)) could overcome permeability defect (quantified in Ussing chambers). Immunofluorescence staining was performed to determine expression of cleaved Notch-1, villin, and claudin 5 in colon samples from mice and humans. Caco-2 cells were infected with a lentivirus containing a specific Notch-1 or scrambled short hairpin RNA sequence. Tight junction assembly was analyzed by immunoblot and immunofluorescence analyses, and transepithelial resistance was monitored. RESULTS: Expression of cleaved Notch-1, villin, or claudin 5 was not detected in RAG1(-/-) colonocytes; their loss correlated with increased intestinal permeability. Transfer of CD45Rb(Hi) and/or CD45Rb(Lo) cells into RAG1(-/-) mice induced expression of cleaved Notch, villin, and claudin 5 in colonocytes and significantly reduced the permeability of the distal colon. Loss of Notch-1 expression in Caco-2 cells correlated with decreased transepithelial resistance and dysregulated expression and localization of tight junction proteins. Levels of cleaved Notch-1 were increased in colonic epithelium of patients with Crohn's disease. CONCLUSIONS: LPLs promote mucosal barrier function, which is associated with activation of the Notch-1 signaling pathway. LPLs maintain intestinal homeostasis by inducing intestinal epithelial cell differentiation, polarization, and barrier function.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Intestinal Mucosa/physiology , Receptor, Notch1/physiology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , Caco-2 Cells , Claudin-5 , Crohn Disease/pathology , Crohn Disease/physiopathology , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , L-Selectin/analysis , L-Selectin/immunology , Leukocyte Common Antigens/analysis , Leukocyte Common Antigens/immunology , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Receptor, Notch1/analysis , Signal Transduction/physiology , Tight Junctions/immunology , Tight Junctions/physiology
3.
J Immunol ; 183(5): 3073-80, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19675158

ABSTRACT

CCR7 is involved in the initiation of immune responses and has been recently implicated in the control of tolerance. To analyze the role of CCR7 in autoimmunity, we backcrossed CCR7(ko/ko) mice (in which ko signifies deficient) onto the autoimmune-prone NOD background. Surprisingly, NODCCR7(ko/ko) mice never developed diabetes, but showed severe inflammation in multiple tissues including thyroid, lung, stomach, intestine, uterus, and testis. NODCCR7(ko/ko) mice had a marked enlargement of the thyroid gland (goiter) that was associated with circulating autoantibodies against thyroglobulin, and development of primary hypothyroidism (decreased levels of serum thyroxin, and augmented levels of thyroid-stimulating hormone in the pituitary gland), features found in Hashimoto's thyroiditis. Cells isolated from diseased thyroids and activated splenocytes from NODCCR7(ko/ko) animals induced goiter in NOD.SCID recipients, demonstrating that autoreactive cells were generated in the absence of CCR7. Moreover, thyroid disease could be accelerated in young NODCCR7(ko/ko) mice by immunization with thyroglobulin. These results demonstrate the complexity in the generation of multiple autoimmune phenotypes in NOD mice and indicate that CCR7 is a key molecule in their development.


Subject(s)
Hypothyroidism/genetics , Hypothyroidism/immunology , Receptors, CCR7/deficiency , Receptors, CCR7/genetics , Thyroiditis, Autoimmune/genetics , Thyroiditis, Autoimmune/immunology , Animals , Cell Movement/genetics , Cell Movement/immunology , Crosses, Genetic , Female , Hypothyroidism/pathology , Immunophenotyping , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Receptors, CCR7/physiology , Thyroiditis, Autoimmune/pathology
4.
J Biol Chem ; 284(36): 24006-16, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19592489

ABSTRACT

Interleukin (IL)-23, a new member of the IL-12 family, plays a central role in the Th17 immune response and in autoimmune diseases. It is clear that activated macrophages and dendritic cells produce IL-23, but the molecular mechanisms whereby inflammatory signals stimulate IL-23 expression are not fully understood. We demonstrate that induction of IL-23 p19 gene expression by LPS depends on the TLR4 and MyD88 pathways. All three MAPK pathways (ERK, JNK, and p38) that are activated by lipopolysaccharide (LPS) stimulation were shown to exert a positive effect on p19 expression. We cloned a 1.3-kb putative p19 promoter and defined its transcription initiation sites by the 5'-rapid amplification of cDNA ends method. By analyzing IL-23 p19 promoter mutants, we have identified a promoter region (-413 to +10) that contains several important elements, including NF-kappaB and AP-1. In addition to NF-kappaB, we have demonstrated that the proximal AP-1 site is important for p19 promoter activation. Mutation of the AP-1 site resulted in the loss of p19 promoter activation. Electrophoretic mobility shift assay (EMSA) analysis showed that c-Jun and c-Fos bind to the AP-1 site, which was confirmed by a chromatin immunoprecipitation assay. Furthermore, co-transfection of c-Jun and ATF2 synergistically induced p19 promoter activation, and c-Jun and ATF2 formed a protein complex, demonstrated by co-immunoprecipitation. Finally, LPS-stimulated peritoneal macrophages from IL-10-deficient mice expressed significantly higher IL-23 p19 than macrophages from wild type mice, and the addition of recombinant IL-10 strongly inhibited LPS-induced p19 expression. Thus, this study suggests that MyD88-dependent Toll-like receptor signaling induces IL-23 p19 gene expression through both MAPKs and NF-kappaB.


Subject(s)
Gene Expression Regulation/physiology , Interleukin-23 Subunit p19/biosynthesis , Macrophages, Peritoneal/metabolism , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 4/metabolism , Transcription Factor AP-1/metabolism , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/immunology , Activating Transcription Factor 2/metabolism , Animals , Autoimmune Diseases , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , Interleukin-10/immunology , Interleukin-10/pharmacology , Interleukin-23 Subunit p19/genetics , Interleukin-23 Subunit p19/immunology , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/immunology , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/immunology , Proto-Oncogene Proteins c-jun/metabolism , Response Elements/genetics , Response Elements/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology
5.
Gastroenterology ; 137(3): 1006-18, 1018.e1-3, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19501588

ABSTRACT

BACKGROUND & AIMS: Chemokines are small proteins that direct leukocyte trafficking under homeostatic and inflammatory conditions. We analyzed the differential expression of chemokines in distinct segments of the intestine and investigated the importance of chemokines for the distribution of leukocytes in the intestine during homeostatic and inflammatory conditions. METHODS: We analyzed messenger RNA for all known chemokines in different segments of the gut by quantitative polymerase chain reaction. To study the effect of multiple-chemokine blockade in the gut, we generated transgenic mice that expressed the chemokine binding protein M3 in the intestine (V-M3 mice). We used flow cytometry to evaluate the changes in the numbers of leukocytes. RESULTS: We observed distinct chemokine expression profiles in the 6 segments of the gut. Some chemokines were expressed throughout the intestine (CCL28, CCL6, CXCL16, and CX3CL1), whereas others were expressed preferentially in the small (CCL25 and CCL5) or large intestine (CCL19, CCL21, and CXCL5). Expression of the chemokine blocker M3 in intestinal epithelial cells resulted in reduced numbers of B and T cells in Peyer's patches, reduced numbers of intraepithelial CD8alphabeta(+)/TCRalphabeta(+) and CD8alphaalpha(+)/TCRalphabeta(+) T cells, and reduced numbers of lamina propria CD8(+) T cells. Strikingly, M3 expression markedly reduced the number of eosinophils and macrophages in the small and large intestines. Dextran sulfate sodium treatment of control mice led to marked changes in the expression of chemokines and in the number of myeloid cells in the colon. These cellular changes were significantly attenuated in the presence of M3. CONCLUSIONS: Our study reveals a complex pattern of chemokine expression in the intestine and indicates that chemokines are critical for leukocyte accumulation in the intestine during homeostasis and inflammation.


Subject(s)
Chemokines/metabolism , Intestines/immunology , Lymphocyte Subsets , Receptors, Chemokine/metabolism , Viral Proteins/metabolism , Animals , In Vitro Techniques , Intestinal Mucosa/metabolism , Mice , Mice, Transgenic , Peyer's Patches/cytology
6.
Gastroenterology ; 135(2): 529-38, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18522803

ABSTRACT

BACKGROUND & AIMS: Several lines of evidence support a role for Toll-like receptor (TLR) signaling to protect the intestine from pathogenic infection. We hypothesized that TLR signaling at the level of the intestinal epithelium is critical for mucosal immune responses. METHODS: We generated transgenic mice that express a constitutively active form of TLR4 in the intestinal epithelium (V-TLR4 mice). Lamina propria cellularity was evaluated by immunostaining and flow cytometry. Immunoglobulin (Ig) A levels in the stool and serum were measured by enzyme-linked immunosorbent assay. Chemokine and cytokine expression were analyzed by quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. RESULTS: V-TLR4 transgenic mice reproduced normally and had a normal life span. Constitutive activity of TLR4 in the intestinal epithelium promoted recruitment of B cells and an increase in fecal IgA levels. Intestinal epithelial cells of V-TLR4 mice expressed higher levels of CCL20 and CCL28, chemokines known to be involved in B-cell recruitment, and of a proliferation-inducing ligand (APRIL), a cytokine that promotes T-cell-independent class switching of B cells to IgA. The changes in B-cell numbers and IgA levels were blocked by simultaneous expression in intestinal epithelial cells of M3, a herpes virus protein that binds and inhibits multiple chemokines. CONCLUSIONS: TLR signaling in the intestinal epithelial cells significantly elevated the production of IgA in the intestine. This effect was mediated by TLR-induced expression of a specific set of chemokines and cytokines that promoted both recruitment of B cells into the lamina propria and IgA class switching of B cells.


Subject(s)
B-Lymphocytes/immunology , Cell Movement , Immunity, Mucosal , Immunoglobulin A/metabolism , Intestinal Mucosa/immunology , Intestine, Small/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/metabolism , Animals , CD4 Antigens/genetics , CD4 Antigens/metabolism , Chemokines/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Feces/chemistry , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Polymerase Chain Reaction , Recombinant Fusion Proteins/metabolism , Toll-Like Receptor 4/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
7.
Eur J Immunol ; 38(7): 1807-13, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18506885

ABSTRACT

IL-17 and IL-22 are typical cytokines produced by the Th17 T cell subset, but it is unclear if Th17 cytokines can be produced by other cell types. We demonstrate that IL-10-deficient and IL-10R-deficient macrophages stimulated with lipopolysaccharide produce high levels of IL-17 and IL-22. Addition of exogenous IL-10 to IL-10-deficient macrophages abolished IL-17 production. When IL-10-deficient and IL-10R-deficient splenocytes were cultured under Th17 polarizing conditions, the population of IL-17-producing cells was increased and the cultures produced significantly higher levels of IL-17 and IL-22. The addition of recombinant IL-10 to IL-10-deficient splenocytes significantly decreased the percentage of IL-17-producing CD4(+) T cells. Finally, the mRNA for the Th17 transcription factor retinoic acid-related orphan receptor (ROR)gammat was significantly elevated in IL-10-deficient spleen cells and macrophages. These data demonstrate that Th17 cytokines and RORgammat are also expressed in macrophages and that IL-10 negatively regulates the expression of Th17 cytokines and RORgammat by both macrophages and T cells.


Subject(s)
Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukins/metabolism , Macrophages/immunology , T-Lymphocyte Subsets/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interleukin-10/deficiency , Interleukin-10/immunology , Interleukin-17/immunology , Interleukins/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Interleukin-10/deficiency , Receptors, Interleukin-10/immunology , Receptors, Interleukin-10/metabolism , Receptors, Retinoic Acid/immunology , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/immunology , Receptors, Thyroid Hormone/metabolism , T-Lymphocyte Subsets/metabolism , Interleukin-22
8.
Eur J Immunol ; 38(5): 1204-14, 2008 May.
Article in English | MEDLINE | ID: mdl-18412165

ABSTRACT

Epstein-Barr virus-induced gene 3 (EBI3) associates with p28 to form IL-27 and with IL-12p35 to form IL-35. IL-27Ralpha(-/-) mice studies indicate that IL-27 negatively regulates Th17 cell differentiation. However, no EBI3, p28 or p35-deficiency studies that directly address the role of EBI3, p28 or p35 on Th17 cells have been done. Here, we demonstrate that spleen cells derived from EBI3(-/-) mice produce significantly higher levels of IL-17 as well as IL-22 upon stimulation with OVA. In vitro derived EBI3(-/-) Th17 cells also produced significantly higher levels of IL-17 and IL-22 than WT cells. The frequency of IL-17-producing cells was also elevated when EBI3(-/-) cells were cultured under Th17 conditions. In addition, spleen cells from EBI3(-/-) mice immunized with Listeria monocytogenes produced significantly elevated levels of IL-17 and IL-22. Furthermore, the Th17 transcription factor RORgamma t was significantly enhanced in EBI3(-/-) cells. Finally, EBI3(-/-) mice exhibited a reduced bacterial load following an acute challenge with L. monocytogenes or a re-challenge of previously immunized mice, suggesting that EBI3 negatively regulates both innate and adaptive immunity. Taken together, these data provide direct evidence that EBI3 negatively regulates the expression of IL-17, IL-22 and RORgamma t as well as protective immunity against L. monocytogenes.


Subject(s)
Gene Expression Regulation , Interleukin-17/metabolism , Interleukins/genetics , Interleukins/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , T-Lymphocytes/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression/genetics , Interferon-gamma/blood , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-17/blood , Interleukin-17/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Minor Histocompatibility Antigens , Nuclear Receptor Subfamily 1, Group F, Member 3 , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Spleen/cytology , Spleen/microbiology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
9.
Cancer Res ; 65(12): 5009-14, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958541

ABSTRACT

The signal pathways that trigger tumor cell escape from immune surveillance are incompletely understood. Toll-like receptors (TLRs), which activate innate and adaptive immune responses, are thought to be restricted to immune cells. We show here that TLRs, including TLR4, are expressed on tumor cells from a wide variety of tissues, suggesting that TLR activation may be an important event in tumor cell immune evasion. Activation of TLR4 signaling in tumor cells by lipopolysaccharide induces the synthesis of various soluble factors and proteins including interleukin-6, inducible nitric oxide synthase, interleukin-12, B7-H1, and B7-H2, and results in resistance of tumor cells to CTL attack. In addition, lipopolysaccharide-stimulated tumor cell supernatants inhibit both T cell proliferation and natural killer cell activity. Blockade of the TLR4 pathway by either TLR4 short interfering RNA or a cell-permeable TLR4 inhibitory peptide reverses tumor-mediated suppression of T cell proliferation and natural killer cell activity in vitro, and in vivo, delays tumor growth and thus prolongs the survival of tumor-bearing mice. These findings indicate that TLR signaling results in a cascade leading to tumor evasion from immune surveillance. These novel functions of TLRs in tumor biology suggest a new class of therapeutic targets for cancer therapy.


Subject(s)
Immunologic Surveillance/immunology , Membrane Glycoproteins/immunology , Neoplasms, Experimental/immunology , Receptors, Cell Surface/immunology , Amino Acid Sequence , Animals , Cell Line, Tumor , Killer Cells, Natural/immunology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Transgenic , Molecular Sequence Data , RNA, Small Interfering/genetics , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Signal Transduction , T-Lymphocytes/immunology , Toll-Like Receptor 4 , Toll-Like Receptors
10.
J Biol Chem ; 280(25): 23531-9, 2005 Jun 24.
Article in English | MEDLINE | ID: mdl-15837792

ABSTRACT

Interferon regulatory factor (IRF)-8/interferon consensus sequence-binding protein is regulated by both transcription and degradation. IRF-8 induced in peritoneal macrophages by interferon-gamma and lipopolysaccharide was degraded rapidly, and degradation of IRF-8 was blocked by MG132, the proteasome inhibitor, but inhibitors of calpain and lysosomal enzymes had no effect. The ubiquitination of IRF-8 was shown by co-immunoprecipitation from RAW264.7 macrophages retrovirally transduced with IRF-8 and hemagglutinin-ubiquitin. The dominant negative ubiquitin mutants K48R and K29R inhibited IRF-8 degradation in 293T cells, confirming the relationship between ubiquitination of IRF-8 and its degradation. IRF-8 carboxyl-terminal truncation mutants were not ubiquitinated and were consequently stable, indicating that the carboxyl-terminal domain of IRF-8 controls ubiquitination. The ubiquitin-protein isopeptide ligase (E3) that ubiquitinated IRF-8 was likely to be Cbl, which formed a complex with IRF-8, demonstrable by both immunoprecipitation and gel filtration. Furthermore, IRF-8 stability was increased by dominant negative Cbl, and IRF-8 ubiquitination was significantly attenuated in Cbl-/- cells. Reflecting increased stability and expression, the IRF-8 carboxyl-terminal deletion mutant induced interleukin (IL)-12 p40 promoter activity much more strongly than IRF-8 did. Furthermore, IRF-8-induced IL-12 p40 synthesis in RAW264.7 cells was enhanced by dominant negative Cbl, and peritoneal macrophages from Cbl-/- mice showed increased IL-12 p40 protein production. Taken together, these results suggest that the proteasomal degradation of IRF-8 mediated by the ubiquitin E3 ligase Cbl down-regulates IL-12 expression.


Subject(s)
Down-Regulation , Interleukin-12/metabolism , Proto-Oncogene Proteins/physiology , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitin/metabolism , Animals , Cell Line , Humans , Hydrolysis , Immunoprecipitation , Interferon Regulatory Factors , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred Strains , Proto-Oncogene Proteins c-cbl
11.
J Biol Chem ; 279(42): 43675-83, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15308666

ABSTRACT

The binding of immune complexes to macrophage Fcgamma receptor results in a subsequent inhibition of lipopolysaccharide-stimulated interleukin-12 synthesis without affecting the induction of tumor necrosis factor-alpha. RNA interference targeting MAST205, a 205-kDa serine/threonine kinase, and transfection of dominant negative MAST205 mutants also mimic this type II macrophage phenotype. Our previous epistasis experiments suggested that the position of MAST205 in the TLR4 signal pathway was proximal to the IkappaB kinase complex. We now report that MAST205 forms a complex with TRAF6, resulting in the inhibition of TRAF6 NF-kappaB activation. We have identified a peptide (residues 218-233) from the N terminus of MAST205 that, when coupled to a protein transduction domain, inhibits the lipopolysaccharide-stimulated activation of NF-kappaB, modulates the size of the MAST205.TRAF6 complex, and inhibits ubiquitination of TRAF6. A dominant negative N-terminal MAST205 deletion mutant also inhibits TRAF6 ubiquitination. The domain required for degradation of MAST205 after Fcgamma receptor activation resides within the N-terminal 261 residues, and degradation is triggered by protein kinase C isoform phosphorylation of Ser/Thr residues. These results suggest that MAST205 functions as a scaffolding protein controlling TRAF6 activity and, therefore, plays an important role in regulating inflammatory responses.


Subject(s)
Microtubule-Associated Proteins/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Factor 6/metabolism , Animals , Cell Line , Enzyme Stability , Macrophages , Mice , NF-kappa B/genetics , Promoter Regions, Genetic , Transfection , Tumor Necrosis Factor-alpha/metabolism
12.
J Immunol ; 172(4): 2559-68, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14764729

ABSTRACT

Stimulation of murine macrophages with LPS results in the coordinated activation of a set of proinflammatory cytokines and costimulatory molecules, including TNF-alpha, IL-6, IL-1, IL-8, IL-12, and CD80. Macrophage LPS-induced synthesis of IL-12 is inhibited following FcgammaR ligation; TNF-alpha secretion is unchanged. We report that microtubule-associated serine/threonine kinase-205 kDa (MAST205) is required for LPS-induced IL-12 synthesis. RNA interference-mediated suppression of MAST205 results in the inhibition of LPS-stimulated IL-12 promoter activity and IL-12 secretion, from both J774 cells and bone marrow-derived macrophages. Similarly, dominant-negative MAST205 mutants inhibit LPS-stimulated IL-12 synthesis and NF-kappaB activation, but do not affect IL-1 or TNF-alpha signaling. Finally, macrophage FcgammaR ligation regulates MAST205 by inducing the rapid ubiquitination and proteasomal degradation of the protein.


Subject(s)
Interleukin-12/biosynthesis , Microtubule-Associated Proteins/physiology , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/physiology , Protein Subunits/biosynthesis , Receptors, IgG/physiology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Cell Line, Tumor , Cells, Cultured , Interleukin-12/antagonists & inhibitors , Interleukin-12 Subunit p40 , Leukemia P388 , Ligands , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/isolation & purification , NF-kappa B/antagonists & inhibitors , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/isolation & purification , Protein Subunits/antagonists & inhibitors , RNA Interference/immunology , Receptors, IgG/immunology , Receptors, IgG/metabolism
13.
J Biol Chem ; 278(4): 2271-7, 2003 Jan 24.
Article in English | MEDLINE | ID: mdl-12429737

ABSTRACT

This study describes the role of the interferon (IFN) consensus sequence-binding protein (ICSBP or IRF-8) in iNOS gene expression by murine macrophages. An ICSBP binding site in the iNOS promoter region (-923 to -913) was identified using an electrophoretic mobility shift assay and chromatin co-immunoprecipitation. Overexpression of ICSBP greatly enhanced IFN-gamma-induced iNOS promoter activation in RAW264.7 cells, and IFN-gamma-induced iNOS promoter activation was abolished in ICSBP-/- macrophages. Furthermore, transduction of retrovirus-ICSBP in ICSBP-/- macrophages rescued IFN-gamma-induced iNOS gene expression. However, transduction of retrovirus-ICSBP in the absence of IFN-gamma activation did not induce iNOS expression in either RAW264.7 cells or ICSBP-/- macrophages. Interestingly, ICSBP alone transduced into ICSBP-/- macrophages did not bind to IFN-stimulated response element site (-923 to -913) of the iNOS promoter region, although following activation with IFN-gamma, a DNA.protein complex was formed that contains ICSBP and IRF-1. Co-transduction of ICSBP with IRF-1 clearly induces nitric oxide production. In addition, interleukin-4 inhibits IFN-gamma-induced iNOS gene expression by attenuating the physical interaction of ICSBP with IRF-1. Complex formation of ICSBP with IRF-1 is essential for iNOS expression, and interleukin-4 attenuates the physical interaction of ICSBP with IRF-1 resulting in the inhibition of INOS gene expression.


Subject(s)
DNA-Binding Proteins/metabolism , Interferon-gamma/metabolism , Interferons/metabolism , Macrophages/metabolism , Nitric Oxide Synthase/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Repressor Proteins/physiology , Animals , Binding Sites , Blotting, Western , Cell Line , DNA/metabolism , Green Fluorescent Proteins , Interferon Regulatory Factor-1 , Interferon Regulatory Factors , Interleukin-4/metabolism , Luminescent Proteins/metabolism , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transduction, Genetic , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...