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










Publication year range
1.
Cell Metab ; 34(6): 874-887.e6, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35504291

ABSTRACT

The tumor microenvironment (TME) contains a rich source of nutrients that sustains cell growth and facilitate tumor development. Glucose and glutamine in the TME are essential for the development and activation of effector T cells that exert antitumor function. Immunotherapy unleashes T cell antitumor function, and although many solid tumors respond well, a significant proportion of patients do not benefit. In patients with KRAS-mutant lung adenocarcinoma, KEAP1 and STK11/Lkb1 co-mutations are associated with impaired response to immunotherapy. To investigate the metabolic and immune microenvironment of KRAS-mutant lung adenocarcinoma, we generated murine models that reflect the KEAP1 and STK11/Lkb1 mutational landscape in these patients. Here, we show increased glutamate abundance in the Lkb1-deficient TME associated with CD8 T cell activation in response to anti-PD1. Combination treatment with the glutaminase inhibitor CB-839 inhibited clonal expansion and activation of CD8 T cells. Thus, glutaminase inhibition negatively impacts CD8 T cells activated by anti-PD1 immunotherapy.


Subject(s)
AMP-Activated Protein Kinase Kinases , Adenocarcinoma of Lung , CD8-Positive T-Lymphocytes , Glutaminase , Lung Neoplasms , AMP-Activated Protein Kinase Kinases/deficiency , AMP-Activated Protein Kinase Kinases/immunology , AMP-Activated Protein Kinase Kinases/metabolism , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Glutaminase/antagonists & inhibitors , Glutaminase/immunology , Humans , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lymphocyte Activation , Mice , Mutation , NF-E2-Related Factor 2/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins p21(ras)/immunology , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Microenvironment
2.
Int J Mol Sci ; 23(2)2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35055042

ABSTRACT

The tumor necrosis factor (TNF) and IL-23/IL-17 axes are the main therapeutic targets in spondyloarthritis. Despite the clinical efficacy of blocking either pathway, monotherapy does not induce remission in all patients and its effect on new bone formation remains unclear. We aimed to study the effect of TNF and IL-17A dual inhibition on clinical disease and structural damage using the HLA-B27/human ß2-microglobulin transgenic rat model of SpA. Immunized rats were randomized according to arthritis severity, 1 week after arthritis incidence reached 50%, to be treated twice weekly for a period of 5 weeks with either a dual blockade therapy of an anti-TNF antibody and an anti-IL-17A antibody, a single therapy of either antibody, or PBS as vehicle control. Treatment-blinded observers assessed inflammation and structural damage clinically, histologically and by micro-CT imaging. Both single therapies as well as TNF and IL-17A dual blockade therapy reduced clinical spondylitis and peripheral arthritis effectively and similarly. Clinical improvement was confirmed for all treatments by a reduction of histological inflammation and pannus formation (p < 0.05) at the caudal spine. All treatments showed an improvement of structural changes at the axial and peripheral joints on micro-CT imaging, with a significant decrease for roughness (p < 0.05), which reflects both erosion and new bone formation, at the level of the caudal spine. The effect of dual blockade therapy on new bone formation was more prominent at the axial than the peripheral level. Collectively, our study showed that dual blockade therapy significantly reduces inflammation and structural changes, including new bone formation. However, we could not confirm a more pronounced effect of dual inhibition compared to single inhibition.


Subject(s)
Interleukin-17/antagonists & inhibitors , Spondylarthritis/etiology , Spondylarthritis/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Arthritis/drug therapy , Arthritis/etiology , Arthritis/metabolism , Arthritis/pathology , Biomarkers , Disease Models, Animal , Disease Susceptibility , Imaging, Three-Dimensional , Immunohistochemistry , Male , Osteogenesis/drug effects , Osteogenesis/genetics , Rats , Rats, Transgenic , Spondylarthritis/diagnosis , Spondylarthritis/drug therapy , X-Ray Microtomography
3.
J Immunol ; 206(5): 1102-1113, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33495237

ABSTRACT

CTLA4-Ig/abatacept dampens activation of naive T cells by blocking costimulation via CD28. It is an approved drug for rheumatoid arthritis but failed to deliver efficacy in a number of other autoimmune diseases. One explanation is that activated T cells rely less on CD28 signaling and use alternate coreceptors for effector function. ICOS is critical for activation of T-dependent humoral immune responses, which drives pathophysiology of IgG-mediated autoimmune diseases. In this study, we asked whether CD28 and ICOS play nonredundant roles for maintenance of T-dependent responses in mouse models. Using a hapten-protein immunization model, we show that during an ongoing germinal center response, combination treatment with CTLA4-Ig and ICOS ligand (ICOSL) blocking Ab completely dissolves ongoing germinal center responses, whereas single agents show only partial activity. Next, we took two approaches to engineer a therapeutic molecule that blocks both pathways. First, we engineered CTLA4-Ig to enhance binding to ICOSL while retaining affinity to CD80/CD86. Using a library approach, binding affinity of CTLA4-Ig to human ICOSL was increased significantly from undetectable to 15-42 nM; however, the affinity was still insufficient to completely block binding of ICOSL to ICOS. Second, we designed a bispecific costimulation inhibitor with high-affinity CTLA4 extracellular domains fused to anti-ICOSL Ab termed bifunctional costimulation inhibitor. With this bispecific approach, we achieved complete inhibition of CD80 and CD86 binding to CD28 as well as ICOS binding to ICOSL. Such bispecific molecules may provide greater therapeutic benefit in IgG-mediated inflammatory diseases compared with CTLA4-Ig alone.


Subject(s)
CD28 Antigens/metabolism , CTLA-4 Antigen/antagonists & inhibitors , Immune Checkpoint Inhibitors/pharmacology , Inducible T-Cell Co-Stimulator Protein/antagonists & inhibitors , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Abatacept/pharmacology , Animals , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , Female , Germinal Center/drug effects , Germinal Center/metabolism , Immunity, Humoral/drug effects , Immunoglobulin G/metabolism , Inducible T-Cell Co-Stimulator Ligand/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes/metabolism
4.
Nature ; 586(7827): 120-126, 2020 10.
Article in English | MEDLINE | ID: mdl-32968282

ABSTRACT

The genetic circuits that allow cancer cells to evade destruction by the host immune system remain poorly understood1-3. Here, to identify a phenotypically robust core set of genes and pathways that enable cancer cells to evade killing mediated by cytotoxic T lymphocytes (CTLs), we performed genome-wide CRISPR screens across a panel of genetically diverse mouse cancer cell lines that were cultured in the presence of CTLs. We identify a core set of 182 genes across these mouse cancer models, the individual perturbation of which increases either the sensitivity or the resistance of cancer cells to CTL-mediated toxicity. Systematic exploration of our dataset using genetic co-similarity reveals the hierarchical and coordinated manner in which genes and pathways act in cancer cells to orchestrate their evasion of CTLs, and shows that discrete functional modules that control the interferon response and tumour necrosis factor (TNF)-induced cytotoxicity are dominant sub-phenotypes. Our data establish a central role for genes that were previously identified as negative regulators of the type-II interferon response (for example, Ptpn2, Socs1 and Adar1) in mediating CTL evasion, and show that the lipid-droplet-related gene Fitm2 is required for maintaining cell fitness after exposure to interferon-γ (IFNγ). In addition, we identify the autophagy pathway as a conserved mediator of the evasion of CTLs by cancer cells, and show that this pathway is required to resist cytotoxicity induced by the cytokines IFNγ and TNF. Through the mapping of cytokine- and CTL-based genetic interactions, together with in vivo CRISPR screens, we show how the pleiotropic effects of autophagy control cancer-cell-intrinsic evasion of killing by CTLs and we highlight the importance of these effects within the tumour microenvironment. Collectively, these data expand our knowledge of the genetic circuits that are involved in the evasion of the immune system by cancer cells, and highlight genetic interactions that contribute to phenotypes associated with escape from killing by CTLs.


Subject(s)
Genome/genetics , Genomics , Neoplasms/genetics , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Escape/genetics , Tumor Escape/immunology , Animals , Autophagy , Cell Line, Tumor , Female , Genes, Neoplasm/genetics , Humans , Interferon-gamma/immunology , Male , Mice , NF-kappa B/metabolism , Reproducibility of Results , Signal Transduction
5.
JCI Insight ; 4(24)2019 12 19.
Article in English | MEDLINE | ID: mdl-31852848

ABSTRACT

Distinct subsets of Tregs reside in nonlymphoid tissues where they mediate unique functions. To interrogate the biology of tissue Tregs in human health and disease, we phenotypically and functionally compared healthy skin Tregs with those in peripheral blood, inflamed psoriatic skin, and metastatic melanoma. The mitochondrial enzyme, arginase 2 (ARG2), was preferentially expressed in Tregs in healthy skin, increased in Tregs in metastatic melanoma, and reduced in Tregs from psoriatic skin. ARG2 enhanced Treg suppressive capacity in vitro and conferred a selective advantage for accumulation in inflamed tissues in vivo. CRISPR-mediated deletion of this gene in primary human Tregs was sufficient to skew away from a tissue Treg transcriptional signature. Notably, the inhibition of ARG2 increased mTOR signaling, whereas the overexpression of this enzyme suppressed it. Taken together, our results suggest that Tregs express ARG2 in human tissues to both regulate inflammation and enhance their metabolic fitness.


Subject(s)
Arginase/metabolism , Skin/pathology , T-Lymphocytes, Regulatory/metabolism , Adoptive Transfer , Adult , Aged , Aged, 80 and over , Animals , Arginase/genetics , Cells, Cultured , Dendritic Cells , Gene Knockout Techniques , Humans , Keratinocytes , Male , Melanoma/immunology , Melanoma/pathology , Mice , Middle Aged , Primary Cell Culture , Psoriasis/immunology , Psoriasis/pathology , RNA-Seq , Signal Transduction/immunology , Skin/cytology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
6.
J Clin Invest ; 129(1): 349-363, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30530991

ABSTRACT

While immune checkpoint blockade leads to potent antitumor efficacy, it also leads to immune-related adverse events in cancer patients. These toxicities stem from systemic immune activation resulting in inflammation of multiple organs, including the gastrointestinal tract, lung, and endocrine organs. We developed a dual variable domain immunoglobulin of anti-CTLA4 antibody (anti-CTLA4 DVD, where CTLA4 is defined as cytotoxic T lymphocyte-associated antigen-4) possessing an outer tumor-specific antigen-binding site engineered to shield the inner anti-CTLA4-binding domain. Upon reaching the tumor, the outer domain was cleaved by membrane type-serine protease 1 (MT-SP1) present in the tumor microenvironment, leading to enhanced localization of CTLA4 blockade. Anti-CTLA4 DVD markedly reduced multiorgan immune toxicity by preserving tissue-resident Tregs in Rag 1-/- mice that received naive donor CD4+ T cells from WT C57BL/6j mice. Moreover, anti-CTLA4 DVD induced potent antitumor effects by decreasing tumor-infiltrating Tregs and increasing the infiltration of antigen-specific CD8+ T lymphocytes in TRAMP-C2-bearing C57BL/6j mice. Treg depletion was mediated through the antibody-dependent cellular cytotoxicity (ADCC) mechanism, as anti-CTLA4 without the FcγR-binding portion (anti-CTLA4 DANA) spared Tregs, preventing treatment-induced toxicities. In summary, our results demonstrate an approach to anti-CTLA4 blockade that depletes tumor-infiltrating, but not tissue-resident, Tregs, preserving antitumor effects while minimizing toxicity. Thus, our tumor-conditional anti-CTLA4 DVD provides an avenue for uncoupling antitumor efficacy from immunotherapy-induced toxicities.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy , Neoplasms/therapy , Single-Chain Antibodies/pharmacology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents, Immunological/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cell Line, Tumor , HEK293 Cells , Humans , Immunity, Cellular , Male , Mice , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Single-Chain Antibodies/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
7.
PLoS One ; 12(8): e0182841, 2017.
Article in English | MEDLINE | ID: mdl-28792532

ABSTRACT

Mutations in the Interleukin (IL)-23/IL-23 receptor loci are associated with increased inflammatory bowel disease (IBD) susceptibility, and IL-23 neutralization has shown efficacy in early clinical trials. To better understand how an excess of IL-23 affects the gastrointestinal tract, we investigated chronic systemic IL-23 exposure in healthy wildtype mice. As expected, IL-23 exposure resulted in early activation of intestinal type 3 innate lymphoid cells (ILC3), followed by infiltration of activated RORγt+ T helper cells. Surprisingly, however, sustained IL-23 stimulus also dramatically reduced classical ILC3 populations within the proximal small intestine, and a phenotypically distinct T-bet expressing ILC3 population emerged. TNFα neutralization, a widely used IBD therapy, reduced several aspects of the IL-23 driven ILC3 response, suggesting a synergy between IL-23 and TNFα in ILC3 activation. In vitro studies supported these findings, revealing previously unappreciated effects of IL-23 and TNFα within the intestine.


Subject(s)
Interleukin-23/metabolism , Intestine, Small/immunology , Lymphocytes/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Female , Flow Cytometry , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Interleukin-23/administration & dosage , Intestine, Small/pathology , Lymphocytes/pathology , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, CCR6/metabolism , Receptors, Interleukin-7/metabolism , Tissue Culture Techniques , Tumor Necrosis Factor-alpha/antagonists & inhibitors
8.
Mol Cancer Ther ; 14(5): 1141-51, 2015 May.
Article in English | MEDLINE | ID: mdl-25731184

ABSTRACT

Despite clinical efficacy, current approved agents targeting EGFR are associated with on-target toxicities as a consequence of disrupting normal EGFR function. MAb 806 is a novel EGFR antibody that selectively targets a tumor-selective epitope suggesting that a mAb 806-based therapeutic would retain antitumor activity without the on-target toxicities associated with EGFR inhibition. To enable clinical development, a humanized variant of mAb 806 designated ABT-806 was generated and is currently in phase 1 trials. We describe the characterization of binding and functional properties of ABT-806 compared with the clinically validated anti-EGFR antibody cetuximab. ABT-806 binds the mutant EGFRvIII with high affinity and, relative to cetuximab, exhibits increased potency against glioblastoma multiforme cell line and patient-derived xenografts expressing this form of the receptor. ABT-806 also inhibits the growth of squamous cell carcinoma xenograft models expressing high levels of wild-type EGFR, associated with inhibition of EGFR signaling, although higher doses of ABT-806 than cetuximab are required for similar activity. ABT-806 enhances in vivo potency of standard-of-care therapies used to treat glioblastoma multiforme and head and neck squamous cell carcinoma. An indium-labeled version of ABT-806, [(111)In]-ABT-806, used to investigate the relationship between dose and receptor occupancy, revealed greater receptor occupancy at lowers doses in an EGFRvIII-expressing model and significant uptake in an orthotopic model. Collectively, these results suggest that ABT-806 may have antitumor activity superior to cetuximab in EGFRvIII-expressing tumors, and similar activity to cetuximab in tumors highly overexpressing wild-type EGFR with reduced toxicity.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Agents/administration & dosage , Cetuximab/administration & dosage , ErbB Receptors/immunology , ErbB Receptors/metabolism , Neoplasms/drug therapy , Animals , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cetuximab/pharmacology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice , Neoplasms/metabolism , Neoplasms/pathology , Protein Binding , Standard of Care , Xenograft Model Antitumor Assays
9.
Cytometry A ; 71(5): 317-26, 2007 May.
Article in English | MEDLINE | ID: mdl-17323353

ABSTRACT

To better define important cell subsets expressing activation markers in rheumatoid arthritis (RA), we compared selective lymphocyte and monocyte B7H1, B7H2, B7RP.1, B7RP.2, and inducible costimulatory molecule (ICOS) expression from normal peripheral blood (NL PB), RA PB, and RA synovial fluid (SF) by multicolor flow cytometry and immunohistochemistry. RA SF memory lymphocytes expressed B7RP.1 and B7RP.2, suggesting that T-cells may function as antigen presenting cells (APCs) in RA joints. We found similar results for ICOS expression. RA SF CD14+ monocytes also expressed B7RP.1 (an ICOS ligand) and the homologous ligand B7RP.2, identifying monocytes as potential mediators of antigen processing and lymphocyte activation in RA. Furthermore, we found an increased population of RA SF CD14+ monocytes expressing B7H1 and B7H2. [The FACS analysis was supported by immunohistochemistry, showing intense lymphocyte and APC (macrophages with dendritic morphology) ICOS staining in RA synovial tissue (ST). Overall, these results define elevated populations of memoryT-lymphocytes expressing proinflammatory B7 molecules in RA SF that either stimulate T cells through ICOS (via ICOS ligands B7RP.1 and B7RP.2), or down-regulate RA ST T-lymphocytes through B7H1 and B7H2.] Therefore, in the same joint, there may exist positive and negative influences on the inflammatory response, and perhaps, the negative signals dominate as joint inflammation resolves.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , B7-1 Antigen/metabolism , Lymphocyte Activation , Lymphocyte Subsets/pathology , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , CD3 Complex/analysis , CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Immunologic Memory , Inducible T-Cell Co-Stimulator Protein , Leukocyte Common Antigens/metabolism , Lipopolysaccharide Receptors/analysis , Lipopolysaccharide Receptors/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/metabolism , Monocytes/metabolism , Synovial Fluid
10.
Brain ; 129(Pt 1): 212-23, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16230319

ABSTRACT

Chemokines and chemokine receptors play a key role in the transmigration of leucocytes across the blood-brain barrier (BBB). CCR2 is the major receptor for CCL2, a potent monocyte and T cell chemoattractant. CCR2 and CCL2 have been consistently associated with a pathogenic role in experimental autoimmune encephalomyelitis, using knockout and transgenic mice, neutralizing antibodies, peptide antagonists and DNA vaccination. However, the significance of CCL2 and CCR2 in multiple sclerosis is enigmatic, because CCL2 levels are consistently decreased in the CSF of patients with this disease and other chronic neuroinflammatory conditions, despite abundant expression within lesional multiple sclerosis tissues. This study used an in vitro BBB model to test the hypothesis that CCL2 is removed from the extracellular fluid by CCR2-positive migrating cells as they cross the BBB, resulting in decreased CSF CCL2 levels. We showed that CCR2-positive T cells and monocytes migrated selectively across the in vitro BBB, and that CCL2 on the abluminal (tissue) side was consumed by migrating T cells and monocytes. Next, we used a new anti-CCR2 antibody to show that CCR2-positive mononuclear inflammatory cells could be readily detected in appropriate positive control tissues, but that CCR2+ cells were very infrequently found in multiple sclerosis lesions. We then showed that CCR2 receptor density on T cells and monocytes was specifically downregulated upon in vitro BBB transmigration in response to CCL2, but not irrelevant chemokines. These findings document a novel strategy for analysing chemokine receptor function in inflammatory CNS disease, and support the hypothesis that CCL2 is consumed by migrating inflammatory cells, which downregulate CCR2, as they cross the BBB.


Subject(s)
Blood-Brain Barrier , Chemokine CCL2/metabolism , Monocytes/metabolism , Multiple Sclerosis/immunology , Receptors, Chemokine/metabolism , T-Lymphocytes/metabolism , Aged , Aged, 80 and over , Cell Movement , Chemokine CCL2/cerebrospinal fluid , Chemotaxis, Leukocyte , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay/methods , Extracellular Fluid/metabolism , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Male , Microscopy, Confocal , Middle Aged , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/metabolism , Pertussis Toxin/pharmacology , Receptors, CCR2
11.
J Immunol ; 173(3): 1779-86, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15265908

ABSTRACT

Previous mouse studies have shown that IL-4 increases the expression of ICOS on activated Th cells, resulting in enhanced ICOS expression on Th2 cells. In this study, we show that ICOS expression on human Th cells is not increased by IL-4, but by IL-12 and by IL-23 instead. Consequently, ICOS expression during IL-12-driven Th1 cell polarization was transiently increased compared with the levels on Th0 cells and IL-4-driven Th2 cells. Addition of IL-12 and/or IL-23 during restimulation increased ICOS expression to the same extent on pre-established Th1, Th2, and Th0 cells, indicating that ICOS levels are not stably imposed by prior polarization. In contrast to the findings in the mouse, IL-4 significantly suppressed the ICOS-enhancing effects of IL-12 and IL-23. The functional consequence of variable ICOS levels was shown in coculture experiments with cells expressing the ICOS-ligand B7-related protein 1 (either transfected Chinese hamster ovary cells or autologous dendritic cells). Ligation of ICOS on 2-day-preactivated effector cells increased their cytokine production to an extent proportional to their ICOS expression levels. As the ICOS-enhancing potentials of IL-12 and IL-23 were maintained for several days after stimulation, both on Th1 and Th2 cells, we propose the concept that local regulation of ICOS expression on activated Th cells by IL-12 and/or IL-23 may provide a powerful means to amplify effector T cell responses in peripheral tissues, independently of the polarized state of the Th cells.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/physiology , Interleukin-12/pharmacology , Interleukins/pharmacology , Lymphocyte Activation , T-Lymphocytes, Helper-Inducer/drug effects , Animals , Antibodies, Monoclonal/immunology , Antigens, Differentiation, T-Lymphocyte/biosynthesis , Antigens, Differentiation, T-Lymphocyte/genetics , B7-1 Antigen/genetics , B7-1 Antigen/immunology , CHO Cells , Coculture Techniques , Cricetinae , Cricetulus , Dendritic Cells/immunology , Female , Gene Expression Regulation/drug effects , Humans , Inducible T-Cell Co-Stimulator Protein , Interleukin-12/antagonists & inhibitors , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukin-4/pharmacology , Interleukins/antagonists & inhibitors , Mice , Mice, Inbred C3H , Recombinant Fusion Proteins/immunology , Recombinant Proteins/pharmacology , Species Specificity , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Transfection
12.
Eur J Immunol ; 34(5): 1282-90, 2004 May.
Article in English | MEDLINE | ID: mdl-15114661

ABSTRACT

The CD28 homologue inducible costimulator (ICOS) has been demonstrated to regulate a number of T cell-dependent immune responses in vivo. However, the expression and functional importance of ICOS during APC-Th cell interaction in the human is not fully understood. Here, we demonstrate that ICOS-mediated signaling plays an important role in the production of selective cytokines during both primary and subsequent Th cell responses upon allospecific or superantigen activation. In contrast, ICOS does not play a role in the differentiation of naive cells into Th1 or Th2 effector cells, nor does it determine the type of effector function of memory cells upon subsequent allogeneic challenge. In addition, our data demonstrate that ICOS provides a novel and unique role in regulating DC-mediated Th2, but not Th1 cell clonal expansion. These data suggest that ICOS-mediated signaling plays a discrete role in the regulation of human T helper cell responses.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Cytokines/biosynthesis , Signal Transduction/physiology , Th2 Cells/metabolism , B7-1 Antigen/metabolism , Cell Differentiation/physiology , Cell Division/physiology , Humans , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Up-Regulation
13.
Immunogenetics ; 53(10-11): 843-50, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11862385

ABSTRACT

The SLAM family of human genes currently consists of seven related members of the immunoglobulin superfamily, membrane-associated proteins, including CD150 (SLAM), CD244 (2B4), CD84, CD229 ( Ly-9), BLAME, CD48, and 19A. These genes are expressed to varying degrees in subsets of immune cells (T, B, natural killer, and myeloid cells) and may function as ligands or receptors. This set of genes, related to CD2 and CD58 on Chromosome (Chr) 1p98, are found clustered close together in the human genome on Chr 1q22. Four of these family members (CD150, CD244, CD84, CD229) contain conserved tyrosine motifs in their cytoplasmic tails that enable them to bind intracellular signaling molecules SAP and EAT-2. SAP is mutated in human X-linked lymphoproliferative disease (XLP), and studies in XLP patients have shown that improper signaling via molecules that bind SAP contributes to the disease. We have identified two new members of the SLAM family (SF), which we term SF2000 and SF2001, which are expressed in immune cells and map in the SLAM gene cluster. SF2001 does not contain SAP-binding motifs in its short cytoplasmic tail. SF2000, which is co-expressed with SAP in T cells, binds both SAP and EAT-2. The data suggest that signaling through SF2000, together with CD150, CD244, CD84, and CD229, is controlled by SAP and therefore contributes to the pathogenesis of XLP.


Subject(s)
CD2 Antigens/chemistry , Glycoproteins/chemistry , Immunoglobulins/chemistry , Intracellular Signaling Peptides and Proteins , Amino Acid Sequence , Animals , Antigens, CD , CD2 Antigens/genetics , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Chromosome Mapping , Cloning, Molecular , Expressed Sequence Tags , Gene Expression Profiling , Glycoproteins/genetics , Humans , Immunoglobulins/genetics , Lymphocytes/metabolism , Molecular Sequence Data , Multigene Family/genetics , Phosphorylation , Phosphotyrosine/metabolism , Phylogeny , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-fyn , RNA, Messenger/metabolism , Receptors, Cell Surface , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , Signaling Lymphocytic Activation Molecule Family Member 1 , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...