Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Nature ; 464(7293): 1362-6, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20200520

ABSTRACT

CD4(+) T helper 2 (T(H)2) cells secrete interleukin (IL)4, IL5 and IL13, and are required for immunity to gastrointestinal helminth infections. However, T(H)2 cells also promote chronic inflammation associated with asthma and allergic disorders. The non-haematopoietic-cell-derived cytokines thymic stromal lymphopoietin, IL33 and IL25 (also known as IL17E) have been implicated in inducing T(H)2 cell-dependent inflammation at mucosal sites, but how these cytokines influence innate immune responses remains poorly defined. Here we show that IL25, a member of the IL17 cytokine family, promotes the accumulation of a lineage-negative (Lin(-)) multipotent progenitor (MPP) cell population in the gut-associated lymphoid tissue that promotes T(H)2 cytokine responses. The IL25-elicited cell population, termed MPP(type2) cells, was defined by the expression of Sca-1 (also known as Ly6a) and intermediate expression of c-Kit (c-Kit(int)), and exhibited multipotent capacity, giving rise to cells of monocyte/macrophage and granulocyte lineages both in vitro and in vivo. Progeny of MPP(type2) cells were competent antigen presenting cells, and adoptive transfer of MPP(type2) cells could promote T(H)2 cytokine responses and confer protective immunity to helminth infection in normally susceptible Il25(-/-) mice. The ability of IL25 to induce the emergence of an MPP(type2) cell population identifies a link between the IL17 cytokine family and extramedullary haematopoiesis, and suggests a previously unrecognized innate immune pathway that promotes T(H)2 cytokine responses at mucosal sites.


Subject(s)
Cell Differentiation , Interleukins/immunology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Antigens, Ly/metabolism , Cell Lineage , Granulocytes/cytology , Granulocytes/immunology , Granulocytes/metabolism , Immunity, Innate/immunology , Immunity, Mucosal/immunology , Interleukins/biosynthesis , Interleukins/metabolism , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Nippostrongylus/immunology , Proto-Oncogene Proteins c-kit/metabolism , Strongylida Infections/immunology , Th2 Cells/cytology , Trichuriasis/immunology , Trichuris/immunology
2.
J Exp Med ; 204(1): 161-70, 2007 Jan 22.
Article in English | MEDLINE | ID: mdl-17200411

ABSTRACT

Interleukin (IL)-25 is a member of the IL-17 family of cytokines. However, unlike the other members of this family, IL-25 promotes T helper (Th) 2 responses. We now show that IL-25 also regulates the development of autoimmune inflammation mediated by IL-17-producing T cells. We have generated IL-25-deficient (il25-/-) mice and found that they are highly susceptible to experimental autoimmune encephalomyelitis (EAE). The accelerated disease in the il25-/- mice is associated with an increase of IL-23 in the periphery and a subsequent increase in the number of inflammatory IL-17-, IFNgamma-, and TNF-producing T cells that invade the central nervous system. Neutralization of IL-17 but not IFNgamma in il25-/- mice prevented EAE, suggesting that IL-17 is a major disease-promoting factor. IL-25 treatment at several time points during a relapse-remitting model or chronic model of EAE completely suppressed disease. IL-25 treatment induced elevated production of IL-13, which is required for suppression of Th17 responses by direct inhibition of IL-23, IL-1beta, and IL-6 expression in activated dendritic cells. Thus, IL-25 and IL-17, being members of the same cytokine family, play opposing roles in the pathogenesis of organ-specific autoimmunity.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/metabolism , Interleukins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Autoimmunity , Base Sequence , Central Nervous System/immunology , DNA/genetics , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Inflammation/etiology , Inflammation/immunology , Interferon-gamma/biosynthesis , Interleukins/deficiency , Interleukins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Th2 Cells/immunology
3.
J Exp Med ; 203(4): 843-9, 2006 Apr 17.
Article in English | MEDLINE | ID: mdl-16606667

ABSTRACT

The cytokine interleukin (IL) 25 has been implicated in the initiation of type 2 immunity by driving the expression of type 2 cytokines such as IL-5 and IL-13, although its role in the regulation of immunity and infection-induced inflammation is unknown. Here, we identify a dual function for IL-25: first, in promoting type 2 cytokine-dependent immunity to gastrointestinal helminth infection and, second, in limiting proinflammatory cytokine production and chronic intestinal inflammation. Treatment of genetically susceptible mice with exogenous IL-25 promoted type 2 cytokine responses and immunity to Trichuris. IL-25 was constitutively expressed by CD4+ and CD8+ T cells in the gut of mouse strains that are resistant to Trichuris, and IL-25-deficient mice on a genetically resistant background failed to develop a type 2 immune response or eradicate infection. Furthermore, chronically infected IL-25(-/-) mice developed severe infection-induced intestinal inflammation associated with heightened expression of interferon-gamma and IL-17, identifying a role for IL-25 in limiting pathologic inflammation at mucosal sites. Therefore, IL-25 is not only a critical mediator of type 2 immunity, but is also required for the regulation of inflammation in the gastrointestinal tract.


Subject(s)
Cytokines/classification , Cytokines/physiology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology , Interleukins/physiology , Trichuriasis/immunology , Animals , Cell Differentiation/immunology , Cells, Cultured , Chronic Disease , Gastrointestinal Tract/parasitology , Inflammation/immunology , Inflammation/parasitology , Inflammation/pathology , Interleukins/genetics , Interleukins/therapeutic use , Mice , Mice, Inbred AKR , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Th1 Cells/cytology , Th1 Cells/immunology , Trichuriasis/drug therapy , Trichuris/immunology
4.
J Exp Med ; 203(12): 2577-87, 2006 Nov 27.
Article in English | MEDLINE | ID: mdl-17074928

ABSTRACT

Aberrant cytokine expression has been proposed as an underlying cause of psoriasis, although it is unclear which cytokines play critical roles. Interleukin (IL)-23 is expressed in human psoriasis and may be a master regulator cytokine. Direct intradermal administration of IL-23 in mouse skin, but not IL-12, initiates a tumor necrosis factor-dependent, but IL-17A-independent, cascade of events resulting in erythema, mixed dermal infiltrate, and epidermal hyperplasia associated with parakeratosis. IL-23 induced IL-19 and IL-24 expression in mouse skin, and both genes were also elevated in human psoriasis. IL-23-dependent epidermal hyperplasia was observed in IL-19-/- and IL-24-/- mice, but was inhibited in IL-20R2-/- mice. These data implicate IL-23 in the pathogenesis of psoriasis and support IL-20R2 as a novel therapeutic target.


Subject(s)
Epidermis/immunology , Epidermis/pathology , Interleukin-23/physiology , Psoriasis/immunology , Psoriasis/pathology , Receptors, Interleukin/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Epidermis/growth & development , Humans , Hyperplasia , Mice , Mice, Knockout , Psoriasis/genetics , RNA, Messenger/biosynthesis , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
5.
Nature ; 442(7101): 461-5, 2006 Jul 27.
Article in English | MEDLINE | ID: mdl-16688182

ABSTRACT

Chronic inflammation has long been associated with increased incidence of malignancy and similarities in the regulatory mechanisms have been suggested for more than a century. Infiltration of innate immune cells, elevated activities of matrix metalloproteases and increased angiogenesis and vasculature density are a few examples of the similarities between chronic and tumour-associated inflammation. Conversely, the elimination of early malignant lesions by immune surveillance, which relies on the cytotoxic activity of tumour-infiltrating T cells or intra-epithelial lymphocytes, is thought to be rate-limiting for the risk to develop cancer. Here we show a molecular connection between the rise in tumour-associated inflammation and a lack of tumour immune surveillance. Expression of the heterodimeric cytokine interleukin (IL)-23, but not of its close relative IL-12, is increased in human tumours. Expression of these cytokines antagonistically regulates local inflammatory responses in the tumour microenvironment and infiltration of intra-epithelial lymphocytes. Whereas IL-12 promotes infiltration of cytotoxic T cells, IL-23 promotes inflammatory responses such as upregulation of the matrix metalloprotease MMP9, and increases angiogenesis but reduces CD8 T-cell infiltration. Genetic deletion or antibody-mediated elimination of IL-23 leads to increased infiltration of cytotoxic T cells into the transformed tissue, rendering a protective effect against chemically induced carcinogenesis. Finally, transplanted tumours are growth-restricted in hosts depleted for IL-23 or in IL-23-receptor-deficient mice. Although many strategies for immune therapy of cancer attempt to stimulate an immune response against solid tumours, infiltration of effector cells into the tumour tissue often appears to be a critical hurdle. We show that IL-23 is an important molecular link between tumour-promoting pro-inflammatory processes and the failure of the adaptive immune surveillance to infiltrate tumours.


Subject(s)
Interleukins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Division , Genetic Predisposition to Disease , Humans , Inflammation/genetics , Inflammation/pathology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukins/biosynthesis , Interleukins/deficiency , Interleukins/genetics , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Neoplasms/immunology , Signal Transduction
6.
J Exp Med ; 201(2): 233-40, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15657292

ABSTRACT

Interleukin (IL)-23 is a heterodimeric cytokine composed of a unique p19 subunit, and a common p40 subunit shared with IL-12. IL-12 is important for the development of T helper (Th)1 cells that are essential for host defense and tumor suppression. In contrast, IL-23 does not promote the development of interferon-gamma-producing Th1 cells, but is one of the essential factors required for the expansion of a pathogenic CD4(+) T cell population, which is characterized by the production of IL-17, IL-17F, IL-6, and tumor necrosis factor. Gene expression analysis of IL-23-driven autoreactive T cells identified a unique expression pattern of proinflammatory cytokines and other novel factors, distinguishing them from IL-12-driven T cells. Using passive transfer studies, we confirm that these IL-23-dependent CD4(+) T cells are highly pathogenic and essential for the establishment of organ-specific inflammation associated with central nervous system autoimmunity.


Subject(s)
Autoimmunity/immunology , CD4-Positive T-Lymphocytes/metabolism , Inflammation/immunology , Interleukins/metabolism , Animals , Autoimmunity/physiology , CD4-Positive T-Lymphocytes/pathology , Central Nervous System/cytology , Central Nervous System/metabolism , Cytokines/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Expression/physiology , Gene Expression Profiling , Inflammation/metabolism , Interleukin-12/metabolism , Interleukin-17/metabolism , Interleukin-23 , Interleukin-23 Subunit p19 , Mice , Time Factors
7.
Int Immunol ; 22(2): 81-90, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19951959

ABSTRACT

IL-23 is a key cytokine in promotion of chronic inflammation. Here, we address if its pro-inflammatory potential can be harnessed to protect against chronic cryptococcosis. Mice were infected with Cryptococcus neoformans and treated with recombinant IL-23. Administration of IL-23 led to prolonged survival and reduced fungal burden but was inferior to IL-12 treatment. Independent of endogenous IL-23/IL-12, IL-23 treatment induced an altered cytokine profile accompanied by marked changes in composition of the inflammatory infiltrate characterized by T cell and dendritic cell recruitment. Although IL-23 induced hallmarks of the T(h)17 pathway, also non-T cells produced IL-17A and IL-22. IL-23 treatment of T-cell-deficient mice resulted in increased IL-17A and IL-22 production and modulation of the cellular response at the site of infection with elevated expression of CD86 on macrophages. Our data show that IL-23 treatment induces innate and adaptive tissue inflammation with limited impact on resistance to chronic cryptococcosis.


Subject(s)
Adaptive Immunity/drug effects , Antifungal Agents/administration & dosage , Cryptococcosis/drug therapy , Cryptococcus neoformans/pathogenicity , Immunity, Innate/drug effects , Interleukin-23/administration & dosage , Animals , Cells, Cultured , Cryptococcosis/immunology , Cryptococcosis/microbiology , Cytokines/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/microbiology , Disease Models, Animal , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inflammation Mediators/metabolism , Interleukin-12/administration & dosage , Interleukin-12/deficiency , Interleukin-12/genetics , Interleukin-12 Subunit p40/deficiency , Interleukin-12 Subunit p40/genetics , Macrophages/drug effects , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/administration & dosage , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/microbiology , Time Factors
8.
J Immunol ; 182(5): 2601-9, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19234154

ABSTRACT

A novel cytokine IL-33, an IL-1 family member, signals via ST2 receptor and promotes Th2 responses, through the activation of NF-kappaB and MAP kinases. Previous studies reported that single Ig IL-1R-related molecule (SIGIRR)/Toll IL-1R8 acts as negative regulator for TLR-IL-1R-mediated signaling. We now found that SIGIRR formed a complex with ST2 upon IL-33 stimulation and specifically inhibited IL-33/ST2-mediated signaling in cell culture model. Furthermore, IL-33-induced Th2 response was enhanced in SIGIRR-deficient mice compared with that in wild-type control mice, suggesting a negative regulatory role of SIGIRR in IL-33/ST2 signaling in vivo. Similar to ST2, SIGIRR was highly expressed in in vitro polarized Th2 cells, but not Th1 cells. SIGIRR-deficient Th2 cells produce higher levels of Th2 cytokines, including IL-5, IL-4, and IL-13, than that in wild-type cells. Moreover, SIGIRR-deficient mice developed stronger Th2 immune response in OVA-challenged asthma model. Taken together, our results suggest that SIGIRR plays an important role in the regulation of Th2 response in vivo, possibly through its impact on IL-33-ST2-mediated signaling.


Subject(s)
Receptors, Interleukin-1/physiology , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Asthma/immunology , Asthma/metabolism , Cell Line , Cells, Cultured , Down-Regulation/immunology , Humans , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/administration & dosage , Interleukins/antagonists & inhibitors , Interleukins/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Interleukin , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Signal Transduction/immunology , Up-Regulation/immunology
9.
J Immunol ; 182(9): 5748-56, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380822

ABSTRACT

IL-27 is secreted by APCs in response to inflammatory stimuli and exerts a proinflammatory Th1-enhancing activity but also has significant anti-inflammatory functions. We examined the molecular mechanism by which IL-27 regulates TGFbeta plus IL-6- or IL-23-dependent Th17 development in the mouse and human systems. IL-27 inhibited the production of IL-17A and IL-17F in naive T cells by suppressing, in a STAT1-dependent manner, the expression of the Th17-specific transcription factor RORgamma t. The in vivo significance of the role of IL-27 was addressed in delayed-type hypersensitivity response and experimental autoimmune encephalomyelitis (EAE). By generating mice deficient for the p28 subunit of IL-27, we showed that IL-27 regulated the severity of delayed-type hypersensitivity response and EAE through its effects on Th17 cells. Furthermore, up-regulation of IL-10 in the CNS, which usually occurs late after EAE onset and plays a role in the resolution of the disease, was notably absent in IL-27p28(-/-) mice. These results show that IL-27 acts as a negative regulator of the developing IL-17A response in vivo, suggesting a potential therapeutic role for IL-27 in autoimmune diseases.


Subject(s)
Cell Lineage/immunology , Growth Inhibitors/physiology , Interleukin-17/antagonists & inhibitors , Interleukins/physiology , Receptors, Retinoic Acid/antagonists & inhibitors , Receptors, Thyroid Hormone/antagonists & inhibitors , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Gene Expression Regulation/immunology , Genetic Predisposition to Disease , Growth Inhibitors/deficiency , Growth Inhibitors/genetics , Humans , Interleukin-17/biosynthesis , Interleukins/deficiency , Interleukins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3 , Protein Subunits/deficiency , Protein Subunits/genetics , Protein Subunits/physiology , Receptors, Retinoic Acid/biosynthesis , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/biosynthesis , Receptors, Thyroid Hormone/genetics , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology
10.
J Exp Med ; 198(12): 1951-7, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14662908

ABSTRACT

Interleukin (IL) 23 is a heterodimeric cytokine composed of a p19 subunit and the p40 subunit of IL-12. IL-23 affects memory T cell and inflammatory macrophage function through engagement of a novel receptor (IL-23R) on these cells. Recent analysis of the contribution of IL-12 and IL-23 to central nervous system autoimmune inflammation demonstrated that IL-23 rather than IL-12 was the essential cytokine. Using gene-targeted mice lacking only IL-12 (p35-/-) or IL-23 (p19-/-), we show that the specific absence of IL-23 is protective, whereas loss of IL-12 exacerbates collagen-induced arthritis. IL-23 gene-targeted mice did not develop clinical signs of disease and were completely resistant to the development of joint and bone pathology. Resistance correlated with an absence of IL-17-producing CD4+ T cells despite normal induction of collagen-specific, interferon-gamma-producing T helper 1 cells. In contrast, IL-12-deficient p35-/- mice developed more IL-17-producing CD4+ T cells, as well as elevated mRNA expression of proinflammatory tumor necrosis factor, IL-1beta, IL-6, and IL-17 in affected tissues of diseased mice. The data presented here indicate that IL-23 is an essential promoter of end-stage joint autoimmune inflammation, whereas IL-12 paradoxically mediates protection from autoimmune inflammation.


Subject(s)
Arthritis, Experimental/etiology , Interleukin-12/physiology , Interleukins/physiology , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/therapy , Collagen Type II/immunology , Female , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Interleukin-23 , Interleukin-23 Subunit p19 , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/biosynthesis
11.
J Immunol ; 181(11): 7891-901, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017979

ABSTRACT

IL-12 is essential for protective T cell-mediated immunity against Salmonella infection. To characterize the role of the related cytokine IL-23, wild-type (WT) C57BL/6 and p19(-/-) mice were infected systemically with an attenuated strain of Salmonella enterica serovar Enteritidis (S. Enteritidis). IL-23-deficient mice controlled infection with S. Enteritidis similarly as WT mice. Similar IFN-gamma production as compared with WT mice, but defective IL-17A and IL-22 production was found in the absence of IL-23. Nevertheless, although IL-23 is required for T cell-dependent cytokine responses, IL-23 is dispensable for protection against S. Enteritidis when IL-12 is present. To analyze the role of IL-23 in the absence of IL-12, low doses of S. Enteritidis were administered to p35(-/-) mice (lacking IL-12), p35/19(-/-) mice (lacking IL-12 and IL-23), p35/40(-/-) mice (lacking IL-12, IL-23, and homodimeric IL-12p40), or p35/IL-17A(-/-) mice (lacking IL-12 and IL-17A). We found survival of p35(-/-) and p35/IL-17A(-/-) mice, whereas p35/19(-/-) and p35/40(-/-) mice died within 3-6 wk and developed liver necrosis. This indicates that IL-23, but not homodimeric IL-12p40, is required for protection, which, surprisingly, is independent of IL-17A. Moreover, protection was associated with IL-22, but not IL-17F or IL-21 expression or with neutrophil recruitment. Finally, anti-IL-22 treatment of S. Enteritidis-infected p35(-/-) mice resulted in liver necrosis, indicating a central role of IL-22 in hepatocyte protection during salmonellosis. In conclusion, IL-23-dependent IL-22, but not IL-17 production is associated with protection against systemic infection with S. Enteritidis in the absence of IL-12.


Subject(s)
Interleukin-12 Subunit p40/immunology , Interleukin-17/immunology , Interleukin-23/immunology , Interleukins/immunology , Salmonella Infections/immunology , Salmonella enteritidis/immunology , Animals , Female , Hepatocytes/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-12 Subunit p40/genetics , Interleukin-17/genetics , Interleukin-23/genetics , Interleukins/genetics , Liver/immunology , Mice , Mice, Knockout , Necrosis/genetics , Necrosis/immunology , Neutrophils/immunology , Salmonella Infections/genetics , Interleukin-22
12.
Mol Cancer Ther ; 19(6): 1298-1307, 2020 06.
Article in English | MEDLINE | ID: mdl-32229606

ABSTRACT

The programmed cell death 1 (PD-1) pathway represents a major immune checkpoint, which may be engaged by cells in the tumor microenvironment to overcome active T-cell immune surveillance. Pembrolizumab (Keytruda®, MK-3475) is a potent and highly selective humanized mAb of the IgG4/kappa isotype designed to directly block the interaction between PD-1 and its ligands, PD-L1 and PD-L2. This blockade enhances the functional activity of T cells to facilitate tumor regression and ultimately immune rejection. Pembrolizumab binds to human and cynomolgus monkey PD-1 with picomolar affinity and blocks the binding of human and cynomolgus monkey PD-1 to PD-L1 and PD-L2 with comparable potency. Pembrolizumab binds both the C'D and FG loops of PD-1. Pembrolizumab overcomes human and cynomolgus monkey PD-L1-mediated immune suppression in T-cell cultures by enhancing IL2 production following staphylococcal enterotoxin B stimulation of healthy donor and cancer patient cells, and IFNγ production in human primary tumor histoculture. Ex vivo and in vitro studies with human and primate T cells show that pembrolizumab enhances antigen-specific T-cell IFNγ and IL2 production. Pembrolizumab does not mediate FcR or complement-driven effector function against PD-1-expressing cells. Pembrolizumab displays dose-dependent clearance and half-life in cynomolgus monkey pharmacokinetic and toxicokinetic studies typical for human IgG4 antibodies. In nonhuman primate toxicology studies, no findings of toxicologic significance were observed. The preclinical data for pembrolizumab are consistent with the clinical anticancer activity and safety that has been demonstrated in human clinical trials.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/pharmacokinetics , Leukocytes, Mononuclear/drug effects , Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , Animals , Antineoplastic Agents, Immunological/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Female , Humans , Immune Checkpoint Inhibitors/pharmacokinetics , Immune Checkpoint Inhibitors/pharmacology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Macaca fascicularis , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , Programmed Cell Death 1 Ligand 2 Protein/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tissue Distribution , Toxicity Tests
13.
J Clin Invest ; 116(5): 1317-26, 2006 May.
Article in English | MEDLINE | ID: mdl-16670771

ABSTRACT

IL-23 is a member of the IL-12 cytokine family that drives a highly pathogenic T cell population involved in the initiation of autoimmune diseases. We have shown that IL-23-dependent, pathogenic T cells produced IL-17 A, IL-17 F, IL-6, and TNF but not IFN-gamma or IL-4. We now show that T-bet and STAT1 transcription factors are not required for the initial production of IL-17. However, optimal IL-17 production in response to IL-23 stimulation appears to require the presence of T-bet. To explore the clinical efficacy of targeting the IL-23 immune pathway, we generated anti-IL-23p19-specific antibodies and tested to determine whether blocking IL-23 function can inhibit EAE, a preclinical animal model of human multiple sclerosis. Anti-IL-23p19 treatment reduced the serum level of IL-17 as well as CNS expression of IFN-gamma, IP-10, IL-17, IL-6, and TNF mRNA. In addition, therapeutic treatment with anti-IL-23p19 during active disease inhibited proteolipid protein (PLP) epitope spreading and prevented subsequent disease relapse. Thus, therapeutic targeting of IL-23 effectively inhibited multiple inflammatory pathways that are critical for driving CNS autoimmune inflammation.


Subject(s)
Autoimmune Diseases/therapy , Encephalomyelitis/therapy , Inflammation/pathology , Interleukins/immunology , Interleukins/physiology , Multiple Sclerosis/therapy , Animals , Chemokine CXCL10 , Chemokines, CXC/metabolism , Disease Models, Animal , Female , Interferon-gamma/metabolism , Interleukin-17/metabolism , Interleukin-23 , Interleukin-23 Subunit p19 , Interleukin-6/metabolism , Interleukins/metabolism , STAT1 Transcription Factor/metabolism , T-Lymphocytes/immunology , Tumor Necrosis Factors/metabolism
14.
J Clin Invest ; 116(5): 1310-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16670770

ABSTRACT

Uncontrolled mucosal immunity in the gastrointestinal tract of humans results in chronic inflammatory bowel disease (IBD), such as Crohn disease and ulcerative colitis. In early clinical trials as well as in animal models, IL-12 has been implicated as a major mediator of these diseases based on the ability of anti-p40 mAb treatment to reverse intestinal inflammation. The cytokine IL-23 shares the same p40 subunit with IL-12, and the anti-p40 mAbs used in human and mouse IBD studies neutralized the activities of both IL-12 and IL-23. IL-10-deficient mice spontaneously develop enterocolitis. To determine how IL-23 contributes to intestinal inflammation, we studied the disease susceptibility in the absence of either IL-23 or IL-12 in this model, as well as the ability of recombinant IL-23 to exacerbate IBD induced by T cell transfer. Our study shows that in these models, IL-23 is essential for manifestation of chronic intestinal inflammation, whereas IL-12 is not. A critical target of IL-23 is a unique subset of tissue-homing memory T cells, which are specifically activated by IL-23 to produce the proinflammatory mediators IL-17 and IL-6. This pathway may be responsible for chronic intestinal inflammation as well as other chronic autoimmune inflammatory diseases.


Subject(s)
Colitis/pathology , Inflammation/pathology , Interleukin-17/physiology , Interleukin-6/physiology , Interleukins/physiology , T-Lymphocytes/pathology , Animals , Autoimmune Diseases/pathology , Humans , Interleukin-23 , Interleukin-23 Subunit p19 , Intestinal Mucosa/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/metabolism
15.
Rheumatology (Oxford) ; 48(12): 1581-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19815670

ABSTRACT

OBJECTIVE: IL-23 is a pro-inflammatory cytokine proposed to be central to the development of autoimmune disease. We investigated whether IL-23, together with the downstream mediator IL-17A, was present and functional in RA in humans. METHODS: RA synovial cells were cultured in the presence or absence of antibodies directed against IL-23p19 or -23R and -17. IL-23, -12, -17, and their receptors, and IL-6, -1beta and TNF-alpha were measured by ELISA and/or PCR. RESULTS: Small amounts of cell-associated IL-23 (median 110 pg/ml) were detected in RA synovial cultures, and found to be functional as IL-23R blockade resulting in a significant inhibition of TNF-alpha (57%), IL-1beta (51%) and IL-6 (30%). However, there was a considerable variability between individual patient samples, and anti-IL-23p19 was found to be considerably less effective. IL-17A protein was detected in approximately 40% of the supernatants and IL-17A blockade, in IL-17A-producing cultures, resulted in a small but significant inhibition of TNF-alpha (38%), IL-1beta (23%) and IL-6 (22%). Addition of recombinant IL-23 to cultures had a variable effect on the spontaneous production of endogenous IL-17A with enhancement observed in some but not all cultures, suggesting that either the low levels of endogenous IL-23 are sufficient to support cytokine production and/or that the relevant Th17 cells were not present. CONCLUSIONS: These results suggest that although IL-23 may have pathogenic activity in a proportion of patients with late-stage RA, it is not abundantly produced in this inflammatory tissue, nor does it have a dominant role in all patient tissues analysed.


Subject(s)
Arthritis, Rheumatoid/immunology , Interleukin-17/immunology , Interleukin-23/immunology , Adult , Aged , Biological Assay/methods , Cells, Cultured , Enzyme-Linked Immunosorbent Assay/methods , Female , Gene Expression , Humans , Inflammation Mediators/metabolism , Interleukin-17/biosynthesis , Interleukin-17/genetics , Interleukin-23/biosynthesis , Interleukin-23/genetics , Male , Middle Aged , RNA, Messenger/genetics , Receptors, Interleukin/biosynthesis , Receptors, Interleukin/genetics , Recombinant Proteins/immunology , Synovial Membrane/immunology
16.
FEMS Immunol Med Microbiol ; 49(3): 353-63, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17378899

ABSTRACT

Lyme borreliosis is characterized by cellular inflammatory responses at multiple body sites. Recently, an association of interleukin-17 (IL-17) and Lyme arthritis was suggested. In this context, it is of special interest that the heterodimeric cytokine IL-23 can act on T cells and initiate the up-regulation of effector cytokines such as IL-17. To determine the role of this specific cytokine cascade for the induction of subsequently induced proinflammatory events we developed an in vitro system to investigate the IL-23-inducing capacity of Borrelia burgdorferi and the potential of the spirochete for inducing the IL-23/IL-17 axis. We used cells derived from mice deficient for IL-23 or IL-12 only or deficient for both IL-12 and IL-23 to define precisely the function of these cytokines. Experiments with bone marrow-derived dendritic cells (BMDC) identified these cells as sources for IL-23 but not for IL-12 after B. burgdorferi exposure. Subsequent investigations with T cell-depleted splenocyte fractions revealed a tight IL-23/IL-17 axis in response to the spirochetes. Monoclonal antibodies that block IL-23 showed further that BMDC-derived IL-23 was required for production of IL-17 in this experimental model. These in vitro data describing a spirochete-induced release of IL-23 may help to define IL-17-dependent inflammatory responses in the disease.


Subject(s)
Borrelia burgdorferi/immunology , Dendritic Cells/immunology , Dendritic Cells/microbiology , Interleukin-17/metabolism , Interleukin-23/biosynthesis , T-Lymphocytes/immunology , Animals , Cells, Cultured , Interleukin-12/deficiency , Interleukin-23/deficiency , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
J Interferon Cytokine Res ; 23(9): 513-22, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14565860

ABSTRACT

Interleukin-27 (IL-27) supports proliferation of naive CD4(+) T cells and enhances interferon-gamma (IFN-gamma) production by activated T cells and natural killer (NK) cells. We report here that IL-27 induces Stat1 and Stat3 phosphorylation and activation in human and murine cell lines and primary human T cells. IL-27 also induces T-Bet, a Stat1-dependent gene crucial to Th1 cell commitment. Similarly, IFN-alpha activates Stat1 and Stat3 and T-Bet expression in naive T cells. Induction of T-Bet results in upregulation of IL-12Rbeta2 on naive T cells, which is essential for responsiveness to IL-12 and differentiation to a Th1 phenotype. Both IL-27 and IFN-alpha induce expression of IL-12Rbeta2 in T cells. In contrast, IFN-gamma, which activates Stat1 but not Stat3, induces expression of T-Bet but not IL-12Rbeta2 in naive T cells. We propose that IL-27 and IFN-alpha are important for early Th1 commitment and act upstream of IL-12 and IFN-gamma in this pathway.


Subject(s)
Interferon-alpha/metabolism , Interleukins/metabolism , Receptors, Interleukin/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Humans , Interferon-alpha/immunology , Interleukins/immunology , Phosphorylation , Receptors, Interleukin-12 , STAT1 Transcription Factor , STAT3 Transcription Factor , T-Box Domain Proteins , Trans-Activators/metabolism
18.
Clin Transl Gastroenterol ; 3: e10, 2012 Feb 16.
Article in English | MEDLINE | ID: mdl-23238132

ABSTRACT

OBJECTIVES: Interleukin-23 (IL-23) has emerged as a new therapeutic target for the treatment of inflammatory bowel disease (IBD). As biomarkers of disease state and treatment efficacy are becoming increasingly important in drug development, we sought to identify efficacy biomarkers for anti-IL-23 therapy in Crohn's disease (CD). METHODS: Candidate IL-23 biomarkers, downstream of IL-23 signaling, were identified using shotgun proteomic analysis of feces and colon lavages obtained from a short-term mouse IBD model (anti-CD40 Rag2(-/-)) treated preventively with monoclonal antibodies (mAbs) to the IL-23 receptor (IL-23R). The biomarkers were then measured in an IBD T-cell transfer model treated therapeutically with a mAb to IL-23 (p19), confirming their association with IBD. To assess the clinical relevance of these markers, we assessed their concentrations in clinical serum, colon tissue, and feces from CD patients. RESULTS: We identified 57 proteins up or downregulated in diseased animals that returned to control values when the mice were treated with mAbs to IL-23R. Among those, S100A8, S100A9, regenerating protein 3ß (REG), REG3γ, lipocalin 2 (LCN2), deleted in malignant tumor 1 (DMBT1), and macrophage migration inhibitory factor (MIF) mRNA levels correlated with disease score and dose titration of mAbs to IL-23R or IL-23(p19). All biomarkers, except DMBT1, were also downregulated after therapeutic administration of mAbs to IL-23(p19) in a T-cell transfer IBD mouse model. In sera from CD patients, we confirmed a significant upregulation of S100A8/A9 (43%), MIF (138%), pancreatitis-associated protein (PAP, human homolog of REG3ß/γ; 49%), LCN2 (520%), and CCL20 (1280%), compared with control samples, as well as a significant upregulation of S100A8/A9 (887%), PAP (401%), and LCN2 (783%) in human feces from CD patients compared with normal controls. CONCLUSIONS: These studies identify multiple protein biomarkers downstream of IL-23 that could be valuable tools to assess the efficacy of this new therapeutic agent.Clinical and Translational Gastroenterology (2012) 3, e10; doi:10.1038/ctg.2012.2; published online 16 February 2012.

19.
Nat Med ; 18(7): 1069-76, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22772566

ABSTRACT

The spondyloarthropathies are a group of rheumatic diseases that are associated with inflammation at anatomically distal sites, particularly the tendon-bone attachments (entheses) and the aortic root. Serum concentrations of interleukin-23 (IL-23) are elevated and polymorphisms in the IL-23 receptor are associated with ankyosing spondylitis, however, it remains unclear whether IL-23 acts locally at the enthesis or distally on circulating cell populations. We show here that IL-23 is essential in enthesitis and acts on previously unidentified IL-23 receptor (IL-23R)(+), RAR-related orphan receptor γt (ROR-γt)(+)CD3(+)CD4(-)CD8(-), stem cell antigen 1 (Sca1)(+) entheseal resident T cells. These cells allow entheses to respond to IL-23 in vitro-in the absence of further cellular recruitment--and to elaborate inflammatory mediators including IL-6, IL-17, IL-22 and chemokine (C-X-C motif) ligand 1 (CXCL1). Notably, the in vivo expression of IL-23 is sufficient to phenocopy the human disease, with the specific and characteristic development of enthesitis and entheseal new bone formation in the initial complete absence of synovitis. As in the human condition, inflammation also develops in vivo at the aortic root and valve, which are structurally similar to entheses. The presence of these entheseal resident cells and their production of IL-22, which activates signal transducer and activator of transcription 3 (STAT3)-dependent osteoblast-mediated bone remodeling, explains why dysregulation of IL-23 results in inflammation at this precise anatomical site.


Subject(s)
Interleukin-23/immunology , Spondylarthropathies/immunology , T-Lymphocytes/immunology , Tendons/immunology , Animals , Antigens, CD/metabolism , Aorta/pathology , Arthritis, Experimental/complications , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Bone Remodeling , CD3 Complex/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Disease Models, Animal , Extremities/pathology , Flow Cytometry , Humans , Immunization, Passive , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Interleukin-17 , Interleukins , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Osteogenesis/immunology , Periosteum/growth & development , Receptors, Interleukin/metabolism , Spondylarthropathies/complications , Spondylarthropathies/pathology , Tendons/pathology , Th17 Cells , Interleukin-22
20.
J Exp Med ; 206(3): 525-34, 2009 Mar 16.
Article in English | MEDLINE | ID: mdl-19273624

ABSTRACT

The C-type lectin-like receptor CD161, which has recently been described to promote T cell expansion, is expressed on a discrete subset of human CD4 T cells. The function of such cells, however, has remained elusive. We now demonstrate that CD161(+) CD4 T cells comprise a circulating and gut-resident T helper 17 (Th17) cell population. During Crohn's disease (CD), these CD161(+) cells display an activated Th17 phenotype, as indicated by increased expression of interleukin (IL)-17, IL-22, and IL-23 receptor. CD161(+) CD4 T cells from CD patients readily produce IL-17 and interferon gamma upon stimulation with IL-23, whereas, in healthy subjects, priming by additional inflammatory stimuli such as IL-1beta was required to enable IL-23-induced cytokine release. Circulating CD161(+) Th17 cells are imprinted for gut homing, as indicated by high levels of CC chemokine receptor 6 and integrin beta7 expression. Supporting their colitogenic phenotype, CD161(+) Th17 cells were found in increased numbers in the inflammatory infiltrate of CD lesions and induced expression of inflammatory mediators by intestinal cells. Our data identify CD161(+) CD4 T cells as a resting Th17 pool that can be activated by IL-23 and mediate destructive tissue inflammation.


Subject(s)
Cell Movement , Inflammation/immunology , Inflammation/pathology , Intestines/immunology , Intestines/pathology , NK Cell Lectin-Like Receptor Subfamily B/immunology , T-Lymphocytes, Helper-Inducer/pathology , Crohn Disease/immunology , Crohn Disease/pathology , Humans , Immunologic Memory/immunology , Interferon-gamma/immunology , Interleukin-17/immunology , Interleukin-23/immunology , Lymphocyte Activation , Organ Specificity , Phenotype , T-Lymphocytes, Helper-Inducer/immunology
SELECTION OF CITATIONS
SEARCH DETAIL