Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Front Immunol ; 7: 4, 2016.
Article in English | MEDLINE | ID: mdl-26834746

ABSTRACT

The SLAMF family (SLAMF) of cell surface glycoproteins is comprised of nine glycoproteins and while SLAMF1, 3, 5, 6, 7, 8, and 9 are self-ligand receptors, SLAMF2 and SLAMF4 interact with each other. Their interactions induce signal transduction networks in trans, thereby shaping immune cell-cell communications. Collectively, these receptors modulate a wide range of functions, such as myeloid cell and lymphocyte development, and T and B cell responses to microbes and parasites. In addition, several SLAMF receptors serve as microbial sensors, which either positively or negatively modulate the function of macrophages, dendritic cells, neutrophils, and NK cells in response to microbial challenges. The SLAMF receptor-microbe interactions contribute both to intracellular microbicidal activity as well as to migration of phagocytes to the site of inflammation. In this review, we describe the current knowledge on how the SLAMF receptors and their specific adapters SLAM-associated protein and EAT-2 regulate innate and adaptive immune responses to microbes.

2.
Tetrahedron ; 72(40): 6078-6083, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-28216796

ABSTRACT

The medical practice for IBD is solely based on anti-inflammatory drugs, but the outcome is far from ideal. Our long-term research goal is to seek a better clinical outcome by combining the anti-inflammatory therapy with physical mucus layer restoration. As the first step towards that objective, we choose to develop self-assembled hydrogels of de novo glycoconjugates that consist of anti-inflammatory drugs and glycopeptides. By covalently linking peptides (e.g., nap-phe-phe-lys), saccharides (e.g., glucosamine), and an anti-inflammatory drug (i.e., olsalazine), we have demonstrated that the obtained molecules self-assemble in water to form hydrogels composed of 3D networks of the nanofibers under acidic conditions. We also confirmed that the resulting glycoconjugates are cell compatible. However, the preliminary assessment of the efficacy of the hydrogels on the murine model is inconclusive, which warrants further investigation and molecular engineering.

3.
Int Immunol ; 27(9): 447-57, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25957267

ABSTRACT

The homophilic cell surface receptors CD150 (Slamf1) and CD352 (Slamf6) are known to modulate adaptive immune responses. Although the Th17 response was enhanced in Slamf6(-/-) C57BL/6 mice upon oral infection with Citrobacter rodentium, the pathologic consequences are indistinguishable from an infection of wild-type C57BL/6 mice. Using a reporter-based binding assay, we show that Slamf6 can engage structures on the outer cell membrane of several Gram(-) bacteria. Therefore, we examined whether Slamf6, like Slamf1, is also involved in innate responses to bacteria and regulates peripheral inflammation by assessing the outcome of C. rodentium infections in Rag(-/-) mice. Surprisingly, the pathology and immune responses in the lamina propria of C. rodentium-infected Slamf6(-/-) Rag(-/-) mice were markedly reduced as compared with those of Rag(-/-) mice. Infiltration of inflammatory phagocytes into the lamina propria was consistently lower in Slamf6(-/-) Rag(-/-) mice than in Rag(-/-) animals. Concomitant with the reduced systemic translocation of the bacteria was an enhanced production of IL-22, suggesting that Slamf6 suppresses a mucosal protective program. Furthermore, administering a mAb (330) that inhibits bacterial interactions with Slamf6 to Rag(-/-) mice ameliorated the infection compared with a control antibody. We conclude that Slamf6-mediated interactions of colonic innate immune cells with specific Gram(-) bacteria reduce mucosal protection and enhance inflammation, contributing to lethal colitis that is caused by C. rodentium infections in Rag(-/-) mice.


Subject(s)
Antigens, CD/immunology , Citrobacter rodentium/immunology , Colitis/immunology , Immunity, Innate/immunology , Receptors, Cell Surface/immunology , Animals , Colitis/microbiology , Colon/immunology , Colon/microbiology , Interleukins/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Signaling Lymphocytic Activation Molecule Family , Signaling Lymphocytic Activation Molecule Family Member 1 , Th17 Cells/immunology , Interleukin-22
4.
Blood ; 125(19): 2937-47, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25833958

ABSTRACT

CD4(+)CD25(+)FoxP3(+) regulatory T cells (Treg) are critical elements for maintaining immune tolerance, for instance to exogenous antigens that are introduced during therapeutic interventions such as cell/organ transplant or gene/protein replacement therapy. Coadministration of antigen with rapamycin simultaneously promotes deletion of conventional CD4(+) T cells and induction of Treg. Here, we report that the cytokine FMS-like receptor tyrosine kinase ligand (Flt3L) enhances the in vivo effect of rapamycin. This occurs via selective expansion of plasmacytoid dendritic cells (pDCs), which further augments the number of Treg. Whereas in conventional DCs, rapamycin effectively blocks mammalian target of rapamycin (mTOR) 1 signaling induced by Flt3L, increased mTOR1 activity renders pDCs more resistant to inhibition by rapamycin. Consequently, Flt3L and rapamycin synergistically promote induction of antigen-specific Treg via selective expansion of pDCs. This concept is supported by the finding that Treg induction is abrogated upon pDC depletion. The combination with pDCs and rapamycin is requisite for Flt3L/antigen-induced Treg induction because Flt3L/antigen by itself fails to induce Treg. As co-administering Flt3L, rapamycin, and antigen blocked CD8(+) T-cell and antibody responses in models of gene and protein therapy, we conclude that the differential effect of rapamycin on DC subsets can be exploited for improved tolerance induction.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Forkhead Transcription Factors/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Membrane Proteins/metabolism , Sirolimus/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Drug Synergism , Flow Cytometry , Humans , Immune Tolerance/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Signal Transduction , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
PLoS One ; 10(3): e0121968, 2015.
Article in English | MEDLINE | ID: mdl-25799045

ABSTRACT

Previous studies have demonstrated that the cell surface receptor Slamf1 (CD150) is requisite for optimal NADPH-oxidase (Nox2) dependent reactive oxygen species (ROS) production by phagocytes in response to Gram- bacteria. By contrast, Slamf8 (CD353) is a negative regulator of ROS in response to Gram+ and Gram- bacteria. Employing in vivo migration after skin sensitization, induction of peritonitis, and repopulation of the small intestine demonstrates that in vivo migration of Slamf1-/- dendritic cells and macrophages is reduced, as compared to wt mice. By contrast, in vivo migration of Slamf8-/- dendritic cells, macrophages and neutrophils is accelerated. These opposing effects of Slamf1 and Slamf8 are cell-intrinsic as judged by in vitro migration in transwell chambers in response to CCL19, CCL21 or CSF-1. Importantly, inhibiting ROS production of Slamf8-/- macrophages by diphenyleneiodonium chloride blocks this in vitro migration. We conclude that Slamf1 and Slamf8 govern ROS-dependent innate immune responses of myeloid cells, thus modulating migration of these cells during inflammation in an opposing manner.


Subject(s)
Antigens, CD/physiology , Myeloid Cells/metabolism , Receptors, Cell Surface/physiology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Movement/genetics , Cell Movement/physiology , Chemotaxis/drug effects , Dendritic Cells/cytology , Dendritic Cells/metabolism , Macrophages/cytology , Macrophages/metabolism , Membrane Proteins , Mice , Mice, Inbred BALB C , Myeloid Cells/cytology , Neutrophils/cytology , Neutrophils/metabolism , Peritonitis/metabolism , Peritonitis/pathology , Reactive Oxygen Species/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
6.
Gastroenterology ; 148(5): 991-1001.e4, 2015 May.
Article in English | MEDLINE | ID: mdl-25678452

ABSTRACT

BACKGROUND & AIMS: Intraepithelial T lymphocyte cells (IEL) are the first immune cells to respond to pathogens; they help maintain the integrity of the epithelial barrier. We studied the function of the mouse glycoprotein Signaling Lymphocyte Activation Molecule Family receptor (SLAMF) 4 (encoded by Slamf4) on the surface of CD8αß αß T-cell receptor (TCR)(+) IELs, and the roles of these cells in homeostasis of the small intestine in mice. METHODS: SLAMF4(-) CD8(+) αßTCR(+) cells isolated from spleens of OT-I Rag1(-/-) mice were induced to express gut-homing receptors and transferred to C57BL/6J mice; levels of SLAMF4(+) cells were measured in small intestine tissues. After administration of anti-CD3 or antigen, with or without anti-SLAM4, to C57BL/6J and Slamf4(-/-) mice, CD8αß αßTCR(+) IELs were collected; cytokine production and cytotoxicity were measured. Depletion of CX3CR1(+) phagocytes was assessed in mice by live-cell confocal imaging or by cytofluorometry; small intestine tissues were analyzed by histology and inflammation was quantified. RESULTS: Splenic CD8(+) αßTCR(+) cells began to express SLAMF4 only after migrating to the small intestine. Injection of C57BL/6J mice with anti-SLAMF4 and anti-CD3 increased levels of interleukin 10 and interferon gamma secretion by IEL, compared with injection of anti-CD3 only. Similarly, the number of granzyme B(+) cytotoxic CD8(+) αßTCR(+) IELs increased in Slamf4(-/-) mice after injection of anti-CD3 and anti-SLAMF4, administration of antigen, or injection of anti-CD3. Surprisingly, in vivo activation of CD8αß(+) IELs with anti-CD3 or antigen caused transient depletion of CX3CR1(+) phagocytes, which was prolonged by co-injection with anti-SLAMF4 or in Slamf4(-/-) mice. Anti-CD3 aggravated inflammation in the small intestines of Slamf4(-/-) mice and Eat2a(-/-)Eat2b(-/-) mice, indicated by flattened villi and crypt hyperplasia. CONCLUSIONS: In mice, the intestinal environment induces SLAMF4 expression and localization to the surface of CD8(+) αßTCR(+) IELs. Signaling via SLAMF4 controls expansion of cytotoxic CD8αß(+) IELs, which regulate the reversible depletion of lamina propria phagocytes and inflammation in the small intestine.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Lymphocyte Activation , Receptors, Immunologic/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD/genetics , CX3C Chemokine Receptor 1 , Cell Movement , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeostasis , Hyperplasia , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/pathology , Mice, Inbred C57BL , Mice, Knockout , Phagocytes/immunology , Phagocytes/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Signal Transduction , Signaling Lymphocytic Activation Molecule Family , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Cytotoxic/immunology
7.
Blood ; 125(15): 2418-27, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25700434

ABSTRACT

Coagulation factor replacement therapy for the X-linked bleeding disorder hemophilia is severely complicated by antibody ("inhibitor") formation. We previously found that oral delivery to hemophilic mice of cholera toxin B subunit-coagulation factor fusion proteins expressed in chloroplasts of transgenic plants suppressed inhibitor formation directed against factors VIII and IX and anaphylaxis against factor IX (FIX). This observation and the relatively high concentration of antigen in the chloroplasts prompted us to evaluate the underlying tolerance mechanisms. The combination of oral delivery of bioencapsulated FIX and intravenous replacement therapy induced a complex, interleukin-10 (IL-10)-dependent, antigen-specific systemic immune suppression of pathogenic antibody formation (immunoglobulin [Ig] 1/inhibitors, IgE) in hemophilia B mice. Tolerance induction was also successful in preimmune mice but required prolonged oral delivery once replacement therapy was resumed. Orally delivered antigen, initially targeted to epithelial cells, was taken up by dendritic cells throughout the small intestine and additionally by F4/80(+) cells in the duodenum. Consistent with the immunomodulatory responses, frequencies of tolerogenic CD103(+) and plasmacytoid dendritic cells were increased. Ultimately, latency-associated peptide expressing CD4(+) regulatory T cells (CD4(+)CD25(-)LAP(+) cells with upregulated IL-10 and transforming growth factor-ß (TGF-ß) expression) as well as conventional CD4(+)CD25(+) regulatory T cells systemically suppressed anti-FIX responses.


Subject(s)
Factor IX/therapeutic use , Hemophilia B/therapy , Administration, Oral , Adoptive Transfer , Animals , Antibody Formation , CD4-Positive T-Lymphocytes/immunology , Factor IX/administration & dosage , Factor IX/genetics , Factor IX/immunology , Hemophilia B/immunology , Humans , Interleukin-10/immunology , Male , Mice , Phytotherapy , Plants, Genetically Modified/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/therapeutic use , Nicotiana/genetics , Transforming Growth Factor beta/immunology
8.
Article in English | MEDLINE | ID: mdl-25364772

ABSTRACT

Adoptive cell therapy utilizing ex vivo expanded polyclonal CD4+CD25+FOXP3+ regulatory T cells (Treg) is in use in clinical trials for the treatment of type 1 diabetes and prevention of graft vs host disease in bone marrow transplantation. Here we seek to evaluate this approach in the treatment of inherited protein deficiencies, i.e. hemophilia, which is often complicated by antibody formation against the therapeutic protein. Treg from mice that express GFP-marked FoxP3 were highly purified by two-step magnetic/flow sorting and ex vivo expanded 50- to 80-fold over a 2-week culture period upon stimulation with antibody-coated microbeads. FoxP3 expression was maintained in >80% of expanded Treg, which also expressed high levels of CD62L and CTLA-4. Transplanted Treg suppressed inhibitory antibody formation against coagulation factors VIII and IX in protein and gene therapies in strain-matched hemophilia A and B mice, including in mice with pre-existing antibodies. Although transplanted Treg became undetectable within two weeks, suppression persisted for >2 months. Additional studies suggested that antigen-specific suppression emerged due to induction of endogenous Treg. The outcomes of these studies support the concept that cell therapy with ex vivo expanded autologous Treg can be used successfully to minimize immune responses in gene and protein replacement therapies.

9.
Front Immunol ; 5: 35, 2014.
Article in English | MEDLINE | ID: mdl-24550919

ABSTRACT

Glucocorticoid-induced tumor necrosis factor receptor family-related protein (TNFRSF18, CD357) is constitutively expressed on regulatory T cells (Tregs) and is inducible on effector T cells. In this report, we examine the role of glucocorticoid-induced TNF receptor family-related protein ligand (GITR-L), which is expressed by antigen presenting cells, on the development and expansion of Tregs. We found that GITR-L is dispensable for the development of naturally occurring FoxP3(+) Treg cells in the thymus. However, the expansion of Treg in GITR-L (-/-) mice is impaired after injection of the dendritic cells (DCs) inducing factor Flt3 ligand. Furthermore, DCs from the liver of GITR-L (-/-) mice were less efficient in inducing proliferation of antigen-specific Treg cells in vitro than the same cells from WT littermates. Upon gene transfer of ovalbumin into hepatocytes of GITR-L (-/-)FoxP3(GFP) reporter mice using adeno-associated virus (AAV8-OVA) the number of antigen-specific Treg in liver and spleen is reduced. The reduced number of Tregs resulted in an increase in the number of ovalbumin specific CD8(+) T effector cells. This is highly significant because proliferation of antigen-specific CD8(+) cells itself is dependent on the presence of GITR-L, as shown by in vitro experiments and by adoptive transfers into GITR-L (-/-) Rag (-/-) and Rag (-/-) mice that had received AAV8-OVA. Surprisingly, administering αCD3 significantly reduced the numbers of FoxP3(+) Treg cells in the liver and spleen of GITR-L (-/-) but not WT mice. Because soluble Fc-GITR-L partially rescues αCD3 induced in vitro depletion of the CD103(+) subset of FoxP3(+)CD4(+) Treg cells, we conclude that expression of GITR-L by antigen presenting cells is requisite for optimal Treg-mediated regulation of immune responses including those in response during gene transfer.

10.
FASEB J ; 28(1): 474-84, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24107315

ABSTRACT

Glucocorticoid-induced TNF receptor family-related protein (GITR) regulates the function of both T cells and antigen-presenting cells (APCs), while the function of GITR ligand (GITR-L) is largely unknown. Here we evaluate the role of GITR-L, whose expression is restricted to APCs, in the development of enterocolitis. On injecting naive CD4(+) T cells, GITR-L(-/-)Rag(-/-) mice develop a markedly milder colitis than Rag(-/-) mice, which correlates with a 50% reduction of Ly6C(+)CD11b(+)MHCII(+) macrophages in the lamina propria and mesenteric lymph nodes. The same result was observed in αCD40-induced acute colitis and during peritonitis, suggesting an altered monocyte migration. In line with these observations, the number of nondifferentiated monocytes was approximately 3-fold higher in the spleen of GITR-L(-/-)Rag(-/-) mice than in Rag(-/-) mice after αCD40 induction. Consistent with the dynamic change in the formation of an active angiotensin II type 1 receptor (AT1) dimer in GITR-L(-/-) splenic monocytes during intestinal inflammation, the migratory capability of splenic monocytes from GITR-L-deficient mice was impaired in an in vitro transwell migration assay. Conversely, αGITR-L reduces the number of splenic Ly6C(hi) monocytes, concomitantly with an increase in AT1 dimers. We conclude that GITR-L regulates the number of proinflammatory macrophages in sites of inflammation by controlling the egress of monocytes from the splenic reservoir.


Subject(s)
Glucocorticoids/pharmacology , Intestinal Mucosa/metabolism , Intestines/immunology , Monocytes/cytology , Receptors, Tumor Necrosis Factor/metabolism , Animals , CD40 Antigens , Mice , Mice, Knockout , Mice, Mutant Strains , Monocytes/drug effects
11.
Adv Drug Deliv Rev ; 65(6): 759-73, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23123293

ABSTRACT

Oral tolerance is defined as the specific suppression of humoral and/or cellular immune responses to an antigen by administration of the same antigen through the oral route. Due to its absence of toxicity, easy administration, and antigen specificity, oral tolerance is a very attractive approach to prevent unwanted immune responses that cause a variety of diseases or that complicate treatment of a disease. Many researchers have induced oral tolerance to efficiently treat autoimmune and inflammatory diseases in different animal models. However, clinical trials yielded limited success. Thus, understanding the mechanisms of oral tolerance induction to therapeutic proteins is critical for paving the way for clinical development of oral tolerance protocols. This review will summarize progress on understanding the major underlying tolerance mechanisms and contributors, including antigen presenting cells, regulatory T cells, cytokines, and signaling pathways. Potential applications, examples for therapeutic proteins and disease targets, and recent developments in delivery methods are discussed.


Subject(s)
Drug Carriers/administration & dosage , Gastrointestinal Tract/immunology , Proteins/administration & dosage , Administration, Oral , Antibody Formation/immunology , Antigen-Presenting Cells/immunology , Cytokines/immunology , Drug Carriers/pharmacokinetics , Humans , Lymphoid Tissue/immunology , Nanoparticles , Plants, Genetically Modified , Recombinant Proteins/administration & dosage , Signal Transduction/immunology , Somatostatin-Secreting Cells/immunology , T-Lymphocytes, Regulatory/immunology
12.
Immunity ; 38(1): 153-65, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23246312

ABSTRACT

Circulatory antigens transit through the small intestine via the fenestrated capillaries in the lamina propria prior to entering into the draining lymphatics. But whether or how this process controls mucosal immune responses remains unknown. Here we demonstrate that dendritic cells (DCs) of the lamina propria can sample and process both circulatory and luminal antigens. Surprisingly, antigen cross-presentation by resident CX3CR1(+) DCs induced differentiation of precursor cells into CD8(+) T cells that expressed interleukin-10 (IL-10), IL-13, and IL-9 and could migrate into adjacent compartments. We conclude that lamina propria CX3CR1(+) DCs facilitate the surveillance of circulatory antigens and act as a conduit for the processing of self- and intestinally absorbed antigens, leading to the induction of CD8(+) T cells, that partake in the control of T cell activation during mucosal immune responses.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Lymphocyte Activation/immunology , Animals , Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CX3C Chemokine Receptor 1 , Cell Differentiation/immunology , Cross-Priming/immunology , Dendritic Cells/metabolism , Enteritis/immunology , Enteritis/prevention & control , Epitopes, T-Lymphocyte/immunology , Intestinal Mucosa/cytology , Intestine, Small/immunology , Mice , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism
13.
Gastroenterology ; 143(6): 1544-1554.e7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22960654

ABSTRACT

BACKGROUND & AIMS: Signaling lymphocyte activation molecule (Slamf)1 is a co-stimulatory receptor on T cells and regulates cytokine production by macrophages and dendritic cells. Slamf1 regulates microbicidal mechanisms in macrophages, therefore we investigated whether the receptor affects development of colitis in mice. METHODS: We transferred CD45RB(hi) CD4(+) T cells into Rag(-/-) or Slamf1(-/-)Rag(-/-) mice to induce colitis. We also induced colitis by injecting mice with an antibody that activates CD40. We determined the severity of enterocolitis based on disease activity index, histology scores, and levels of cytokine production, and assessed the effects of antibodies against Slamf1 on colitis induction. We quantified migration of monocytes and macrophage to inflamed tissues upon induction of colitis or thioglycollate-induced peritonitis and in response to tumor necrosis factor-α in an air-pouch model of leukocyte migration. RESULTS: Colitis was reduced in Slamf1(-/-)Rag(-/-) mice, compared with Rag(-/-) mice, after transfer of CD45RB(hi) CD4(+) T cells or administration of the CD40 agonist. The numbers of monocytes and macrophages were reduced in inflamed tissues of Slamf1(-/-)Rag(-/-) mice, compared with Rag(-/-) mice, after induction of colitis and other inflammatory disorders. An antibody that inhibited Slamf1 reduced the level of enterocolitis in Rag(-/-) mice. CONCLUSIONS: Slamf1 contributes to the development of colitis in mice. It appears to indirectly regulate the appearance of monocytes and macrophages in inflamed intestinal tissues. Antibodies that inhibit Slamf1 reduce colitis in mice, so human SLAMF1 might be a therapeutic target for inflammatory bowel disease.


Subject(s)
Antigens, CD/physiology , Colitis/physiopathology , Receptors, Cell Surface/physiology , Animals , Antigens, CD/genetics , CD40 Antigens/adverse effects , Cell Movement , Chemokine CCL2/blood , Chemokine CCL7/blood , Colitis/blood , Colitis/chemically induced , Disease Models, Animal , Intestines/pathology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Signaling Lymphocytic Activation Molecule Family Member 1
14.
J Immunol ; 188(12): 5829-32, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22593622

ABSTRACT

Slamf8 (CD353) is a cell surface receptor that is expressed upon activation of macrophages (MΦs) by IFN-γ or bacteria. In this article, we report that a very high NADPH oxidase (Nox2) enzyme activity was found in Slamf8(-/-) MΦs in response to Escherichia coli or Staphylococcus aureus, as well as to PMA. The elevated Nox2 activity in Slamf8(-/-) MΦs was also demonstrated in E. coli or S. aureus phagosomes by using a pH indicator system and was further confirmed by a reduction in the enzyme activity after transfection of the receptor into Slamf8-deficient primary MΦs or RAW 264.7 cells. Upon exposure to bacteria or PMA, protein kinase C activity in Slamf8(-/-) MΦs is increased. This results in an enhanced phosphorylation of p40phox, one key component of the Nox2 enzyme complex, which, in turn, leads to greater Nox2 activity. Taken together, the data show that, in response to inflammation-associated stimuli, the inducible receptor Slamf8 negatively regulates inflammatory responses.


Subject(s)
Antigens, CD/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Receptors, Cell Surface/metabolism , Animals , Antigens, CD/immunology , Blotting, Western , Cell Line , Gene Expression Regulation/immunology , Inflammation/immunology , Inflammation/metabolism , Macrophages/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/immunology , Phagosomes/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Cell Surface/immunology , Signaling Lymphocytic Activation Molecule Family Member 1 , Up-Regulation
15.
Gastroenterology ; 142(3): 582-591.e8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22155173

ABSTRACT

BACKGROUND & AIMS: The glucocorticoid-induced tumor necrosis factor receptor family-related protein (GITR; also called TNFRSF18 or CD357) regulates the T cell-mediated immune response and is present on surfaces of regulatory T (Treg) cells and activated CD4(+) T cells. We investigated the roles of GITR in the development of colitis in mice. METHODS: Chronic enterocolitis was induced by the transfer of wild-type or GITR(-/-) CD4(+) T cells to GITR(-/-) × Rag(-/-) or Rag(-/-) mice. We determined the severity of colitis by using the disease activity index; measured levels of inflammatory cytokines, T cells, and dendritic cells; and performed histologic analysis of colon samples. RESULTS: Transfer of nonfractionated CD4(+) cells from wild-type or GITR(-/-) donors induced colitis in GITR(-/-) × Rag(-/-) but not in Rag(-/-) mice. Among mice with transfer-induced colitis, the percentage of Treg and T-helper (Th) 17 cells was reduced but that of Th1 cells increased. Treg cells failed to prevent colitis in GITR(-/-) × Rag(-/-) recipients; this was not the result of aberrant function of GITR(-/-) Treg or T effector cells but resulted from an imbalance between the numbers of tolerogenic CD103(+) and PDCA1(+) plasmacytoid dendritic cells in GITR(-/-) mice. This imbalance impaired Treg cell development and expanded the Th1 population in GITR(-/-) × Rag(-/-) mice following transfer of nonfractionated CD4(+) cells. CONCLUSIONS: GITR is not required on the surface of Treg and T effector cells to induce colitis in mice; interactions between GITR and its ligand are not required for induction of colitis. GITR instead appears to control dendritic cell and monocyte development; in its absence, mice develop aggravated chronic enterocolitis via an imbalance of colitogenic Th1 cells and Treg cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Colon/immunology , Glucocorticoid-Induced TNFR-Related Protein/metabolism , T-Lymphocyte Subsets/immunology , Animals , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/transplantation , Cell Proliferation , Cells, Cultured , Chronic Disease , Colitis/genetics , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Cytokines/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Glucocorticoid-Induced TNFR-Related Protein/deficiency , Glucocorticoid-Induced TNFR-Related Protein/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inflammation Mediators/metabolism , Integrin alpha Chains/metabolism , Intestinal Mucosa/immunology , Ligands , Lymph Nodes/immunology , Lymphocyte Transfusion , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Severity of Illness Index , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Time Factors
16.
Mol Ther ; 19(7): 1263-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21386826

ABSTRACT

Hepatic gene transfer using adeno-associated viral (AAV) vectors has been shown to efficiently induce immunological tolerance to a variety of proteins. Regulatory T-cells (Treg) induced by this route suppress humoral and cellular immune responses against the transgene product. In this study, we examined the roles of immune suppressive cytokines interleukin-10 (IL-10) and transforming growth factor-ß (TGF-ß) in the development of tolerance to human coagulation factor IX (hF.IX). Interestingly, IL-10 deficient C57BL/6 mice receiving gene transfer remained tolerant to hF.IX and generated Treg that suppressed anti-hF.IX formation. Effects of TGF-ß blockade were also minor in this strain. In contrast, in C3H/HeJ mice, a strain known to have stronger T-cell responses against hF.IX, IL-10 was specifically required for the suppression of CD8(+) T-cell infiltration of the liver. Furthermore, TGF-ß was critical for tipping the balance toward an regulatory immune response. TGF-ß was required for CD4(+)CD25(+)FoxP3(+) Treg induction, which was necessary for suppression of effector CD4(+) and CD8(+) T-cell responses as well as antibody formation. These results demonstrate the crucial, nonredundant roles of IL-10 and TGF-ß in prevention of immune responses against AAV-F.IX-transduced hepatocytes.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/genetics , Interleukin-10/metabolism , Liver/metabolism , Transforming Growth Factor beta/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Enzyme-Linked Immunosorbent Assay , Factor IX , Flow Cytometry , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Hepatocytes/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-10/genetics , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Mice , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/antagonists & inhibitors
17.
J Exp Med ; 208(4): 811-22, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-21422172

ABSTRACT

Studies of human systemic lupus erythematosus patients and of murine congenic mouse strains associate genes in a DNA segment on chromosome 1 with a genetic predisposition for this disease. The systematic analysis of lupus-prone congenic mouse strains suggests a role for two isoforms of the Ly108 receptor in the pathogenesis of the disease. In this study, we demonstrate that Ly108 is involved in the pathogenesis of lupus-related autoimmunity in mice. More importantly, we identified a third protein isoform, Ly108-H1, which is absent in two lupus-prone congenic animals. Introduction of an Ly108-H1-expressing transgene markedly diminishes T cell-dependent autoimmunity in congenic B6.Sle1b mice. Thus, an immune response-suppressing isoform of Ly108 can regulate the pathogenesis of lupus.


Subject(s)
Antigens, Ly/genetics , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/prevention & control , Animals , Autoimmunity , Base Sequence , CD4-Positive T-Lymphocytes/immunology , Exons , Female , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Isoforms/genetics
18.
Nat Immunol ; 11(10): 920-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20818396

ABSTRACT

Phagocytosis is a pivotal process by which macrophages eliminate microorganisms after recognition by pathogen sensors. Here we unexpectedly found that the self ligand and cell surface receptor SLAM functioned not only as a costimulatory molecule but also as a microbial sensor that controlled the killing of gram-negative bacteria by macrophages. SLAM regulated activity of the NADPH oxidase NOX2 complex and phagolysosomal maturation after entering the phagosome, following interaction with the bacterial outer membrane proteins OmpC and OmpF. SLAM recruited a complex containing the intracellular class III phosphatidylinositol kinase Vps34, its regulatory protein kinase Vps15 and the autophagy-associated molecule beclin-1 to the phagosome, which was responsible for inducing the accumulation of phosphatidylinositol-3-phosphate, a regulator of both NOX2 function and phagosomal or endosomal fusion. Thus, SLAM connects the gram-negative bacterial phagosome to ubiquitous cellular machinery responsible for the control of bacterial killing.


Subject(s)
Antigens, CD/metabolism , Escherichia coli Infections/immunology , Escherichia coli/immunology , Macrophages/immunology , Phagosomes/immunology , Receptors, Cell Surface/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Animals , Antigens, CD/genetics , Apoptosis Regulatory Proteins/metabolism , Bacterial Proteins/genetics , Beclin-1 , Cells, Cultured , Endosomal Sorting Complexes Required for Transport/metabolism , Macrophages/microbiology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Chaperones/genetics , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Phagocytosis , Phagosomes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Porins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/genetics , Signaling Lymphocytic Activation Molecule Family Member 1 , Vacuolar Sorting Protein VPS15
19.
Int Immunol ; 22(4): 259-70, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20139172

ABSTRACT

Naturally occurring regulatory T cells (Treg) express high levels of glucocorticoid-induced tumour necrosis factor receptor (GITR). However, studies of the role of GITR in Treg biology has been complicated by the observation that upon activation effector CD4(+) T (Teff) cells also express the receptor. Here, we dissect the contribution of GITR-induced signaling networks in the expansion and function of FoxP3(+) Treg. We demonstrate that a high-affinity soluble Fc-GITR-L dimer, in conjugation with alphaCD3, specifically enhances in vitro proliferation of Treg, which retain their phenotypic markers (CD25 and FoxP3) and their suppressor function, while minimally affecting Teff cells. Furthermore, Fc-GITR-L does not impair Teff susceptibility to suppression, as judged by cocultures employing GITR-deficient and GITR-sufficient CD4(+) T-cell subsets. Notably, this expansion of Treg could also be seen in vivo, by injecting FoxP3-IRES-GFP mice with Fc-GITR-L even in the absence of antigenic stimulation. In order to test the efficacy of these findings therapeutically, we made use of a C3H/HeJ hemophilia B-prone mouse model. The use of liver-targeted human coagulation factor IX (hF.IX) gene therapy in this model has been shown to induce liver toxicity and the subsequent failure of hF.IX expression. Interestingly, injection of Fc-GITR-L into the hemophilia-prone mice that were undergoing liver-targeted hF.IX gene therapy increased the expression of F.IX and reduced the anticoagulation factors. We conclude that GITR engagement enhances Treg proliferation both in vitro and in vivo and that Fc-GITR-L may be a useful tool for in vivo tolerance induction.


Subject(s)
Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cell Proliferation , Disease Models, Animal , Factor IX/genetics , Forkhead Transcription Factors/metabolism , Genetic Therapy , Glucocorticoid-Induced TNFR-Related Protein , Hemophilia B/therapy , Humans , Immune Tolerance , Immunoglobulin Fc Fragments/administration & dosage , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Ligands , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Receptors, Nerve Growth Factor/administration & dosage , Receptors, Nerve Growth Factor/genetics , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
20.
PLoS One ; 4(8): e6376, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19652717

ABSTRACT

BACKGROUND: Hepatic gene transfer, in particular using adeno-associated viral (AAV) vectors, has been shown to induce immune tolerance to several protein antigens. This approach has been exploited in animal models of inherited protein deficiency for systemic delivery of therapeutic proteins. Adequate levels of transgene expression in hepatocytes induce a suppressive T cell response, thereby promoting immune tolerance. This study addresses the question of whether AAV gene transfer can induce tolerance to a cytoplasmic protein. MAJOR FINDINGS: AAV-2 vector-mediated hepatic gene transfer for expression of cytoplasmic beta-galactosidase (beta-gal) was performed in immune competent mice, followed by a secondary beta-gal gene transfer with E1/E3-deleted adenoviral Ad-LacZ vector to provoke a severe immunotoxic response. Transgene expression from the AAV-2 vector in approximately 2% of hepatocytes almost completely protected from inflammatory T cell responses against beta-gal, eliminated antibody formation, and significantly reduced adenovirus-induced hepatotoxicity. Consequently, approximately 10% of hepatocytes continued to express beta-gal 45 days after secondary Ad-LacZ gene transfer, a time point when control mice had lost all Ad-LacZ derived expression. Suppression of inflammatory T cell infiltration in the liver and liver damage was linked to specific transgene expression and was not seen for secondary gene transfer with Ad-GFP. A combination of adoptive transfer studies and flow cytometric analyses demonstrated induction of Treg that actively suppressed CD8(+) T cell responses to beta-gal and that was amplified in liver and spleen upon secondary Ad-LacZ gene transfer. CONCLUSIONS: These data demonstrate that tolerance induction by hepatic AAV gene transfer does not require systemic delivery of the transgene product and that expression of a cytoplasmic neo-antigen in few hepatocytes can induce Treg and provide long-term suppression of inflammatory responses and immunotoxicity.


Subject(s)
Cytoplasm/enzymology , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Immune Tolerance/genetics , Liver/metabolism , beta-Galactosidase/immunology , Animals , Liver/enzymology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transduction, Genetic , beta-Galactosidase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL