Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Immunol ; 20(9): 1138-1149, 2019 09.
Article in English | MEDLINE | ID: mdl-31427775

ABSTRACT

Interleukin (IL)-1R3 is the co-receptor in three signaling pathways that involve six cytokines of the IL-1 family (IL-1α, IL-1ß, IL-33, IL-36α, IL-36ß and IL-36γ). In many diseases, multiple cytokines contribute to disease pathogenesis. For example, in asthma, both IL-33 and IL-1 are of major importance, as are IL-36 and IL-1 in psoriasis. We developed a blocking monoclonal antibody (mAb) to human IL-1R3 (MAB-hR3) and demonstrate here that this antibody specifically inhibits signaling via IL-1, IL-33 and IL-36 in vitro. Also, in three distinct in vivo models of disease (crystal-induced peritonitis, allergic airway inflammation and psoriasis), we found that targeting IL-1R3 with a single mAb to mouse IL-1R3 (MAB-mR3) significantly attenuated heterogeneous cytokine-driven inflammation and disease severity. We conclude that in diseases driven by multiple cytokines, a single antagonistic agent such as a mAb to IL-1R3 is a therapeutic option with considerable translational benefit.


Subject(s)
Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors , Peritonitis/immunology , Pneumonia/immunology , Psoriasis/immunology , A549 Cells , Animals , Cell Line, Tumor , Disease Models, Animal , HEK293 Cells , Humans , Imiquimod/toxicity , Inflammation/pathology , Interleukin-1/immunology , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1beta/immunology , Interleukin-33/immunology , Male , Mice , Mice, Inbred C57BL , Ovalbumin/toxicity , Peritonitis/drug therapy , Peritonitis/pathology , Pneumonia/drug therapy , Pneumonia/pathology , Psoriasis/drug therapy , Psoriasis/pathology , Signal Transduction/immunology , Uric Acid/toxicity
2.
Blood ; 129(17): 2384-2394, 2017 04 27.
Article in English | MEDLINE | ID: mdl-28122740

ABSTRACT

Understanding leukemia heterogeneity is critical for the development of curative treatments as the failure to eliminate therapy-persistent leukemic stem cells (LSCs) may result in disease relapse. Here we have combined high-throughput immunophenotypic screens with large-scale single-cell gene expression analysis to define the heterogeneity within the LSC population in chronic phase chronic myeloid leukemia (CML) patients at diagnosis and following conventional tyrosine kinase inhibitor (TKI) treatment. Our results reveal substantial heterogeneity within the putative LSC population in CML at diagnosis and demonstrate differences in response to subsequent TKI treatment between distinct subpopulations. Importantly, LSC subpopulations with myeloid and proliferative molecular signatures are proportionally reduced at a higher extent in response to TKI therapy compared with subfractions displaying primitive and quiescent signatures. Additionally, cell surface expression of the CML stem cell markers CD25, CD26, and IL1RAP is high in all subpopulations at diagnosis but downregulated and unevenly distributed across subpopulations in response to TKI treatment. The most TKI-insensitive cells of the LSC compartment can be captured within the CD45RA- fraction and further defined as positive for CD26 in combination with an aberrant lack of cKIT expression. Together, our results expose a considerable heterogeneity of the CML stem cell population and propose a Lin-CD34+CD38-/lowCD45RA-cKIT-CD26+ population as a potential therapeutic target for improved therapy response.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/therapeutic use , Single-Cell Analysis/methods , ADP-ribosyl Cyclase 1/deficiency , ADP-ribosyl Cyclase 1/genetics , ADP-ribosyl Cyclase 1/immunology , Antigens, CD34/genetics , Antigens, CD34/immunology , Biomarkers, Tumor/immunology , Case-Control Studies , Cell Lineage/immunology , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/immunology , Gene Expression , Genetic Heterogeneity , Humans , Immunophenotyping , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukocyte Common Antigens/deficiency , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/immunology , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-kit/deficiency , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/immunology , Treatment Outcome
3.
Proc Natl Acad Sci U S A ; 112(34): 10786-91, 2015 Aug 25.
Article in English | MEDLINE | ID: mdl-26261316

ABSTRACT

Acute myeloid leukemia (AML) is associated with a poor survival rate, and there is an urgent need for novel and more efficient therapies, ideally targeting AML stem cells that are essential for maintaining the disease. The interleukin 1 receptor accessory protein (IL1RAP; IL1R3) is expressed on candidate leukemic stem cells in the majority of AML patients, but not on normal hematopoietic stem cells. We show here that monoclonal antibodies targeting IL1RAP have strong antileukemic effects in xenograft models of human AML. We demonstrate that effector-cell-mediated killing is essential for the observed therapeutic effects and that natural killer cells constitute a critical human effector cell type. Because IL-1 signaling is important for the growth of AML cells, we generated an IL1RAP-targeting antibody capable of blocking IL-1 signaling and show that this antibody suppresses the proliferation of primary human AML cells. Hence, IL1RAP can be efficiently targeted with an anti-IL1RAP antibody capable of both achieving antibody-dependent cellular cytotoxicity and blocking of IL-1 signaling as modes of action. Collectively, these results provide important evidence in support of IL1RAP as a target for antibody-based treatment of AML.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Animals , Antibody-Dependent Cell Cytotoxicity , Cell Division , Cell Line, Tumor , Cytotoxicity, Immunologic , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1 Receptor Accessory Protein/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Proteins/immunology , Xenograft Model Antitumor Assays
4.
Blood ; 121(18): 3709-13, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23479569

ABSTRACT

IL1RAP, a co-receptor for interleukin (IL)-1 and IL-33 receptors, was previously found to be highly upregulated on candidate chronic myeloid leukemia stem cells, allowing for leukemia-selective killing using IL1RAP-targeting antibodies. We analyzed IL1RAP expression in a consecutive series of 29 patients with acute myeloid leukemia (AML) and, based on the level of expression in mononuclear cells (MNCs), we divided the samples into 3 groups: IL1RAP low (n = 6), IL1RAP intermediate (n = 11), and IL1RAP high (n = 12). Within the CD34+CD38- population, the intermediate and high groups expressed higher levels of IL1RAP than did corresponding normal cells. With the aim to target AML stem cells, an anti-IL1RAP monoclonal antibody was generated followed by isotype switching for improved antibody-dependent, cell-mediated cytotoxicity activity. Using this antibody, we achieved selective killing of AML MNC, CD34+CD38+, and CD34+CD38- cells. Our findings demonstrate that IL1RAP is a promising new therapeutic target in AML.


Subject(s)
Antibodies/pharmacology , Cytotoxicity, Immunologic/drug effects , Interleukin-1 Receptor Accessory Protein/immunology , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/immunology , Adult , Aged , Aged, 80 and over , Antibody Specificity , Cells, Cultured , Female , Humans , Immunotherapy/methods , Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors , Male , Middle Aged , Molecular Targeted Therapy , Neoplastic Stem Cells/pathology
5.
Proc Natl Acad Sci U S A ; 109(35): 14128-33, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22904187

ABSTRACT

The proinflammatory cytokine interleukin-1 (IL-1) signals via type I IL-1 receptor (IL-1RI) and IL-1 receptor accessory protein (IL1RAP), which leads to activation of the transcription factor NF-κB and induction of a range of downstream proteins involved in inflammatory and immune responses. Here, we identified the E3 ubiquitin ligase membrane-associated RING-CH (MARCH8) as a suppressor of IL-1ß-induced NF-κB- and MAPK-activation pathways. Overexpression of MARCH8 inhibits IL-1ß-induced NF-κB and MAPK activation, whereas knockdown of MARCH8 has the opposite effect. Mechanistically, MARCH8 interacts with IL1RAP and targets its Lys512 for K48-linked polyubiquitination and degradation. Our findings suggest that MARCH8-mediated polyubiquitination and degradation of IL1RAP is an important mechanism for negative regulation of IL-1ß-induced signaling pathways.


Subject(s)
Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1beta/immunology , MAP Kinase Signaling System/immunology , NF-kappa B/immunology , Ubiquitin-Protein Ligases/immunology , Gene Expression/immunology , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Inflammation/immunology , Inflammation/metabolism , Interleukin-1 Receptor Accessory Protein/metabolism , Interleukin-1beta/metabolism , Membrane Proteins/genetics , Membrane Proteins/immunology , Membrane Proteins/metabolism , NF-kappa B/metabolism , Polyubiquitin/immunology , Polyubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/immunology
6.
J Allergy Clin Immunol ; 134(1): 170-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24568840

ABSTRACT

BACKGROUND: Genome-wide association studies identified IL33 and IL-1 receptor-like 1 (IL1RL1)/IL18R1 as asthma susceptibility loci. IL33 and IL1RL1 constitute a single ligand-receptor pathway. OBJECTIVE: In 2 birth cohorts, the Prevalence and Incidence of Asthma and Mite Allergy (PIAMA) study and Avon Longitudinal Study of Parents and Children (ALSPAC), we analyzed associations of longitudinal wheezing phenotypes and asthma with single nucleotide polymorphisms (SNPs) of 8 genes encoding IL-33, IL1RL1, its coreceptor IL1RAcP, its adaptors myeloid differentiation primary response gene 88 (MyD88) and Toll-IL-11 receptor domain containing adaptor protein (TIRAP), and the downstream IL-1 receptor-associated kinase 1, IL-1 receptor-associated kinase 4, and TNF receptor-associated factor 6 (TRAF6). Furthermore, we investigated whether SNPs in this pathway show replicable evidence of gene-gene interaction. METHODS: Ninety-four SNPs were investigated in 2007 children in the PIAMA study and 7247 children in ALSPAC. Associations with wheezing phenotypes and asthma at 8 years of age were analyzed in each cohort and subsequently meta-analyzed. Gene-gene interactions were assessed through model-based multifactor dimensionality reduction in the PIAMA study, and gene-gene interactions of 10 SNP pairs were further evaluated. RESULTS: Intermediate-onset wheeze was associated with SNPs in several genes in the IL33-IL1RL1 pathway after applying multiple testing correction in the meta-analysis: 2 IL33 SNPs (rs4742170 and rs7037276), 1 IL-1 receptor accessory protein (IL1RAP) SNP (rs10513854), and 1 TRAF6 SNP (rs5030411). Late-onset wheeze was associated with 2 IL1RL1 SNPs (rs10208293 and rs13424006), and persistent wheeze was associated with 1 IL33 SNP (rs1342326) and 1 IL1RAP SNP (rs9290936). IL33 and IL1RL1 SNPs were nominally associated with asthma. Three SNP pairs showed interaction for asthma in the PIAMA study but not in ALSPAC. CONCLUSIONS: IL33-IL1RL1 pathway polymorphisms are associated with asthma and specific wheezing phenotypes; that is, most SNPs are associated with intermediate-onset wheeze, a phenotype closely associated with sensitization. We speculate that IL33-IL1RL1 pathway polymorphisms affect development of wheeze and subsequent asthma through sensitization in early childhood.


Subject(s)
Asthma/genetics , Genetic Predisposition to Disease , Interleukins/genetics , Polymorphism, Single Nucleotide , Receptors, Cell Surface/genetics , Respiratory Sounds/physiopathology , Asthma/immunology , Asthma/pathology , Child , Child, Preschool , Cohort Studies , Epistasis, Genetic , Female , Humans , Infant , Infant, Newborn , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-33 , Interleukins/immunology , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Receptors, Cell Surface/immunology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Respiratory Sounds/immunology , Signal Transduction
7.
Proc Natl Acad Sci U S A ; 107(37): 16280-5, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805474

ABSTRACT

Chronic myeloid leukemia (CML) is genetically characterized by the Philadelphia (Ph) chromosome, formed through a reciprocal translocation between chromosomes 9 and 22 and giving rise to the constitutively active tyrosine kinase P210 BCR/ABL1. Therapeutic strategies aiming for a cure of CML will require full eradication of Ph chromosome-positive (Ph(+)) CML stem cells. Here we used gene-expression profiling to identify IL-1 receptor accessory protein (IL1RAP) as up-regulated in CML CD34(+) cells and also in cord blood CD34(+) cells as a consequence of retroviral BCR/ABL1 expression. To test whether IL1RAP expression distinguishes normal (Ph(-)) and leukemic (Ph(+)) cells within the CML CD34(+)CD38(-) cell compartment, we established a unique protocol for conducting FISH on small numbers of sorted cells. By using this method, we sorted cells directly into drops on slides to investigate their Ph-chromosome status. Interestingly, we found that the CML CD34(+)CD38(-)IL1RAP(+) cells were Ph(+), whereas CML CD34(+)CD38(-)IL1RAP(-) cells were almost exclusively Ph(-). By performing long-term culture-initiating cell assays on the two cell populations, we found that Ph(+) and Ph(-) candidate CML stem cells could be prospectively separated. In addition, by generating an anti-IL1RAP antibody, we provide proof of concept that IL1RAP can be used as a target on CML CD34(+)CD38(-) cells to induce antibody-dependent cell-mediated cytotoxicity. This study thus identifies IL1RAP as a unique cell surface biomarker distinguishing Ph(+) from Ph(-) candidate CML stem cells and opens up a previously unexplored avenue for therapy of CML.


Subject(s)
Antibodies/immunology , Apoptosis , Interleukin-1 Receptor Accessory Protein/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , ADP-ribosyl Cyclase 1/immunology , Antigens, CD34/immunology , Cell Separation , Fusion Proteins, bcr-abl/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Interleukin-1 Receptor Accessory Protein/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Membrane Glycoproteins/immunology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/immunology
8.
Biochem Biophys Res Commun ; 417(4): 1310-5, 2012 Jan 27.
Article in English | MEDLINE | ID: mdl-22234307

ABSTRACT

The hepatitis B virus (HBV) Precore protein is processed through the secretory pathway directly as HBeAg or with the generation of an intermediate (P20). Precore gene has been shown to be implicated in viral persistence, but the functions of HBeAg and its precursors have not been fully elucidated. We show that the secreted proteins HBeAg and P20 interact with T cell surface and alter Kit-225 and primary T cells proliferation, a process which may facilitate the establishment of HBV persistence. Our data indicate that the N-terminal end of Precore is important for these inhibitory effects and exclude that they are dependent on the association of HBeAg and P20 with two characterized cell surface ligands, the Interleukin-1 Receptor Accessory Protein and gC1qR (present study).


Subject(s)
Cell Proliferation , Hepatitis B e Antigens/immunology , Hepatitis B virus/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , T-Lymphocytes/virology , Amino Acid Sequence , Carrier Proteins/immunology , Cells, Cultured , Hepatitis B e Antigens/chemistry , Hepatitis B e Antigens/genetics , Humans , Interleukin-1 Receptor Accessory Protein/immunology , Mitochondrial Proteins/immunology , Molecular Sequence Data
9.
J Clin Immunol ; 31(3): 455-64, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21181432

ABSTRACT

INTRODUCTION: Interleukin (IL)-1 is a key orchestrator of inflammation and IL-1 inhibitors are expected to be promising pharmaceutical agents for such pathologies. IL-1 is bound to the complex of two receptor components with much higher affinity than with either receptor component alone. MATERIALS AND METHODS: We examined the effect of a heterodimer of IL-1 receptor accessory protein (Acp)-immunoglobulin (Ig) and IL-1R type II (IL1R2)-Ig named AcP-Ig/IL1R2-Ig heterodimer, and compared its effects with other IL-1 inhibitors reported previously. RESULTS AND DISCUSSION: Our results demonstrated that the rat AcP-Ig/IL1R2-Ig heterodimer (IC50=1.95 pM) inhibited IL-1 response to a greater extent than IL1RA (IC50=1,935 pM), Acp-IL1R type I (IL1R1)-Ig homodimer (IC50=73.7 pM) and Acp-IL1R2-Ig homodimer (IC50=72.8 pM). Moreover, human AcP-Ig/IL1R2-Ig heterodimer (IC50=0.14 pM) inhibited it to a greater extent than Acp-IL1R1-Ig homodimer (IC50=4.48 pM) and strongly inhibited responses of both IL-1α and IL-1ß. CONCLUSIONS: The AcP-Ig/IL1R2-Ig heterodimer, which is similar to the original extracellular structure of the Acp/IL1R1 complex, may inhibit the IL-1 response more vigorously than other IL-1 blocking biopharmaceutical agents.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Fibroblasts/drug effects , Fibroblasts/immunology , Immunoglobulins/metabolism , Interleukin-1 Receptor Accessory Protein/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Multiprotein Complexes/pharmacology , Receptors, Interleukin-1/metabolism , Recombinant Proteins/metabolism , Animals , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Base Sequence , COS Cells , Cells, Cultured , Chlorocebus aethiops , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Inflammation , Inhibitory Concentration 50 , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Molecular Sequence Data , Multiprotein Complexes/immunology , Multiprotein Complexes/metabolism , Plasmids , Protein Binding , Protein Engineering , Protein Multimerization , Rats , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Receptors, Interleukin-1 Type II/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Transfection
10.
Front Immunol ; 12: 779100, 2021.
Article in English | MEDLINE | ID: mdl-35003094

ABSTRACT

Interleukin-1 (IL-1) family cytokines are potent mediators of inflammation, acting to coordinate local and systemic immune responses to a wide range of stimuli. Aberrant signaling by IL-1 family cytokine members, however, is linked to myriad inflammatory syndromes, autoimmune conditions and cancers. As such, blocking the inflammatory signals inherent to IL-1 family signaling is an established and expanding therapeutic strategy. While several FDA-approved IL-1 inhibitors exist, including an Fc fusion protein, a neutralizing antibody, and an antagonist cytokine, none specifically targets the co-receptor IL-1 receptor accessory protein (IL-1RAcP). Most IL-1 family cytokines form productive signaling complexes by binding first to their cognate receptors - IL-1RI for IL-1α and IL-1ß; ST2 for IL-33; and IL-36R for IL-36α, IL-36ß and IL-36γ - after which they recruit the shared secondary receptor IL-1RAcP to form a ternary cytokine/receptor/co-receptor complex. Recently, IL-1RAcP was identified as a biomarker for both AML and CML. IL-1RAcP has also been implicated in tumor progression in solid tumors and an anti-IL1RAP antibody (nadunolimab, CAN04) is in phase II clinical studies in pancreatic cancer and non-small cell lung cancer (NCT03267316). As IL-1RAcP is common to all of the abovementioned IL-1 family cytokines, targeting this co-receptor raises the possibility of selective signaling inhibition for different IL-1 family cytokines. Indeed, previous studies of IL-1ß and IL-33 signaling complexes have revealed that these cytokines employ distinct mechanisms of IL-1RAcP recruitment even though their overall cytokine/receptor/co-receptor complexes are structurally similar. Here, using functional, biophysical, and structural analyses, we show that antibodies specific for IL-1RAcP can differentially block signaling by IL-1 family cytokines depending on the distinct IL-1RAcP epitopes that they engage. Our results indicate that targeting a shared cytokine receptor is a viable therapeutic strategy for selective cytokine signaling inhibition.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antibodies/pharmacology , Epitopes , Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors , Interleukin-1beta/metabolism , Interleukin-33/metabolism , Anti-Inflammatory Agents/immunology , Anti-Inflammatory Agents/metabolism , Antibodies/immunology , Antibodies/metabolism , Antibody Affinity , Antibody Specificity , Binding Sites, Antibody , HEK293 Cells , Humans , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1 Receptor Accessory Protein/metabolism , Molecular Docking Simulation , Molecular Targeted Therapy , Protein Binding , Signal Transduction
11.
Front Immunol ; 10: 1412, 2019.
Article in English | MEDLINE | ID: mdl-31281320

ABSTRACT

Interleukin-1 (IL-1) family cytokines are key signaling molecules in both the innate and adaptive immune systems, mediating inflammation in response to a wide range of stimuli. The basic mechanism of signal initiation is a stepwise process in which an agonist cytokine binds its cognate receptor. Together, this cytokine-receptor complex recruits an often-common secondary receptor. Intracellularly, the Toll/IL-1 Receptor (TIR) domains of the two receptors are brought into close proximity, initiating an NF-κB signal transduction cascade. Due to the potent inflammatory response invoked by IL-1 family cytokines, several physiological mechanisms exist to inhibit IL-1 family signaling, including antagonist cytokines and decoy receptors. The numerous cytokines and receptors in the IL-1 superfamily are further classified into four subfamilies, dependent on their distinct cognate receptors-the IL-1, IL-33, and IL-36 subfamilies share IL-1RAcP as their secondary receptor, while IL-18 subfamily utilizes a distinct secondary receptor. Here, we describe how structural biology has informed our understanding of IL-1 family cytokine signaling, with a particular focus on molecular mechanisms of signaling complex formation and antagonism at the atomic level, as well as how these findings have advanced therapeutics to treat some chronic inflammatory diseases that are the result of dysregulated IL-1 signaling.


Subject(s)
Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1/immunology , NF-kappa B/immunology , Signal Transduction/immunology , Humans
12.
Cancer Res ; 79(3): 663-675, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30514753

ABSTRACT

Chronic myeloid leukemia (CML) is a chronic disease resulting in myeloid cell expansion through expression of the BCR-ABL1 fusion transcript. Tyrosine kinase inhibitors (TKI) have significantly increased survival of patients with CML, and deep responders may consider stopping the treatment. However, more than 50% of patients relapse and restart TKI, subsequently suffering unknown toxicity. Because CML is a model immune system-sensitive disease, we hypothesize that chimeric antigen receptor (CAR) T cells targeting IL1 receptor-associated protein (IL1RAP) in quiescent CML stem cells may offer an opportunity for a permanent cure. In this study, we produced and molecularly characterized a specific monoclonal anti-IL1RAP antibody from which fragment antigen-binding nucleotide coding sequences were cloned as a single chain into a lentiviral backbone and secured with the suicide gene iCASP9/rimiducid system. Our CAR T-cell therapy exhibited cytotoxicity against both leukemic stem cells and, to a lesser extent, monocytes expressing IL1RAP, with no apparent effect on the hematopoietic system, including CD34+ stem cells. This suggests IL1RAP as a tumor-associated antigen for immunotherapy cell targeting. IL1RAP CAR T cells were activated in the presence of IL1RAP+ cell lines or primary CML cells, resulting in secretion of proinflammatory cytokines and specifically killing in vitro and in a xenograft murine model. Overall, we demonstrate the proof of concept of a CAR T-cell immunotherapy approach in the context of CML that is applicable for young patients and primary TKI-resistant, intolerant, or allograft candidate patients. SIGNIFICANCE: These findings present the first characterization and proof of concept of a chimeric antigen receptor directed against IL1RAP expressed by leukemic stem cells in the context of CML.


Subject(s)
Hematopoietic Stem Cells/immunology , Immunotherapy, Adoptive/methods , Interleukin-1 Receptor Accessory Protein/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/transplantation , Animals , Antibodies, Monoclonal/immunology , Cell Engineering/methods , Cytotoxicity, Immunologic , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Inbred BALB C , Molecular Targeted Therapy , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
14.
Biomed Pharmacother ; 107: 1064-1073, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30257318

ABSTRACT

High expression of Interluekin-1 receptor accessory protein chain (IL-1RAcP) and activated IL-1 signaling were found in some tumor types. IL-1RAcP is considered as the common accessory chain in the IL-1R family, and it is essential for the initiation of IL-1 signaling of all the receptor complexes that encompass it. Thus, the selection and characterization of human anti- IL-1RAcP single-chain antibody fragments variable (scFv) is the first step toward the construction of new anticancer monoclonal antibodies designed for optimal cancer therapy. Here, we found that IL-1RAcP expression was increased in both triple-negative breast cancer (TNBC) cell line cells and TNBC patient cohort, and correlated with shorter recurrence-free survival (RFS). In this study, we employed a human scFv-displaying phage library for the first establishment an antagonistic anti-IL-1RAcP human antibody, scFv 12H7. scFv 12H7 was found a high affinity and specificity binder of IL-1RAcP by a series assays, including EC50, IC50,KD values test and cell binding determination by flow cytometry and immunofluorescence. Also, scFv 12H7 was demonstrated bearing growth inhibitory activity of TNBC cells in vitro and in vivo. Mechanisms study showed that IL-1-activated-NF-κB pathway was significantly inhibited in TNBC cells by incubation with scFv 7H12 for 24 h. Crystal structure analysis, mutations introduction, and yeast two-hybrid assay showed that scFv 12H7 interacted with residues in the D1-D2 domain of IL-1RAcP, which further indicated that scFv 12H7 was a functional binding to IL-1RAcP and uncovered its structure mechanism. In conclusion, scFv 12H7 represent excellent therapeutic candidates for further preclinical and clinical development of TNBC therapy.


Subject(s)
Antibodies, Monoclonal/immunology , Interleukin-1 Receptor Accessory Protein/immunology , Single-Chain Antibodies/immunology , Triple Negative Breast Neoplasms/therapy , Animals , Antibodies, Monoclonal/administration & dosage , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/metabolism , Signal Transduction/immunology , Triple Negative Breast Neoplasms/immunology , Xenograft Model Antitumor Assays
15.
J Biochem Mol Biol ; 40(4): 562-70, 2007 Jul 31.
Article in English | MEDLINE | ID: mdl-17669273

ABSTRACT

Soluble or cell-bound IL-1 receptor accessory protein (IL-1RAcP) does not bind IL-1 but rather forms a complex with IL-1 and IL-1 receptor type I (IL-1RI) resulting in signal transduction. Synthetic peptides to various regions in the Ig-like domains of IL-1RAcP were used to produce antibodies and these antibodies were affinity-purified using the respective antigens. An anti-peptide-4 antibody which targets domain III inhibited 70% of IL-1beta-induced productions of IL-6 and PGE(2) from 3T3-L1 cells. Anti-peptide-2 or 3 also inhibited IL-1-induced IL-6 production by 30%. However, anti-peptide-1 which is directed against domain I had no effect. The antibody was more effective against IL-1beta compared to IL-1alpha. IL-1-induced IL-6 production was augmented by coincubation with PGE(2). The COX inhibitor ibuprofen blocked IL-1-induced IL-6 and PGE(2) production. These results confirm that IL-1RAcP is essential for IL-1 signaling and that increased production of IL-6 by IL-1 needs the co-induction of PGE(2). However, the effect of PGE(2) is independent of expressions of IL-1RI and IL-1RAcP. Our data suggest that domain III of IL-1RAcP may be involved in the formation or stabilization of the IL-1RI/IL-1 complex by binding to epitopes on domain III of the IL-1RI created following IL-1 binding to the IL-1RI.


Subject(s)
Antibodies/pharmacology , Dinoprostone/biosynthesis , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1alpha/pharmacology , Interleukin-1beta/pharmacology , Interleukin-6/biosynthesis , 3T3-L1 Cells , Animals , Cells, Cultured , Dinoprostone/pharmacology , Down-Regulation/drug effects , Humans , Ibuprofen/pharmacology , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/metabolism , Mice , Peptides/immunology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism
16.
Mol Immunol ; 87: 308-316, 2017 07.
Article in English | MEDLINE | ID: mdl-28531815

ABSTRACT

The use of cytokines as adjuvants in poultry is promising because they may enhance immune responses to antigens. In this study, we created two mutants, chicken interleukin-1 beta (ChIL-1ß) Q19A and R140A, which exhibited significantly increased in vivo biological activity compared with wild-type ChIL-1ß. The potential mucosal adjuvant activity of the mutants Q19A and R140A was evaluated in chickens through the intranasal coadministration of a single dose of the Newcastle disease virus (NDV) vaccine with Q19A or R140A. Compared with chickens vaccinated with only the NDV vaccine or the NDV vaccine plus wild-type recombinant ChIL-1ß, chickens vaccinated with Q19A or R140A had significantly increased serum hemagglutination-inhibition antibody titers and anti-NDV-specific IgA antibody levels 1 week later, a high amount of interferon-γ secretion from splenocytes, and increased secretory IgA accumulated in nasal tissues. In addition, molecular dynamics simulations of the mutant R140A bound to its receptor (IL-1RI) and receptor accessory protein (IL-1RAcP) were more energetically favorable than the analogous wild-type ternary complex resulting in a decreased energy, which may stabilize the R140A/IL-1RI/IL-1RAcP complex. In conclusion, the mutants Q19A and R140A are effective adjuvants that accelerate and enhance chicken mucosal immunity when co-administered with one dose of the NDV vaccine.


Subject(s)
Chickens/immunology , Immunity, Mucosal/immunology , Interleukin-1beta/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Immunoglobulin A/immunology , Interferon-gamma/immunology , Interleukin-1 Receptor Accessory Protein/immunology , Newcastle Disease/immunology , Newcastle disease virus/immunology , Receptors, Interleukin-1 Type I/immunology , Vaccination/methods
17.
Mucosal Immunol ; 9(5): 1173-82, 2016 09.
Article in English | MEDLINE | ID: mdl-26813341

ABSTRACT

We previously reported that TLR4(-/-) mice are refractory to mouse-adapted A/PR/8/34 (PR8) influenza-induced lethality and that therapeutic administration of the TLR4 antagonist Eritoran blocked PR8-induced lethality and acute lung injury (ALI) when given starting 2 days post infection. Herein we extend these findings: anti-TLR4- or -TLR2-specific IgG therapy also conferred significant protection of wild-type (WT) mice from lethal PR8 infection. If treatment is initiated 3 h before PR8 infection and continued daily for 4 days, Eritoran failed to protect WT and TLR4(-/-) mice, implying that Eritoran must block a virus-induced, non-TLR4 signal that is required for protection. Mechanistically, we determined that (i) Eritoran blocks high-mobility group B1 (HMGB1)-mediated, TLR4-dependent signaling in vitro and circulating HMGB1 in vivo, and an HMGB1 inhibitor protects against PR8; (ii) Eritoran inhibits pulmonary lung edema associated with ALI; (iii) interleukin (IL)-1ß contributes significantly to PR8-induced lethality, as evidenced by partial protection by IL-1 receptor antagonist (IL-1Ra) therapy. Synergistic protection against PR8-induced lethality was achieved when Eritoran and the antiviral drug oseltamivir were administered starting 4 days post infection. Eritoran treatment does not prevent development of an adaptive immune response to subsequent PR8 challenge. Overall, our data support the potential of a host-targeted therapeutic approach to influenza infection.


Subject(s)
Acute Lung Injury/drug therapy , Antiviral Agents/pharmacology , Disaccharides/pharmacology , Immunoglobulin G/pharmacology , Orthomyxoviridae Infections/drug therapy , Oseltamivir/pharmacology , Sugar Phosphates/pharmacology , Acute Lung Injury/immunology , Acute Lung Injury/mortality , Acute Lung Injury/virology , Animals , Drug Synergism , Female , Gene Expression Regulation , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/genetics , HMGB1 Protein/immunology , Immunity, Innate , Interleukin-1 Receptor Accessory Protein/antagonists & inhibitors , Interleukin-1 Receptor Accessory Protein/genetics , Interleukin-1 Receptor Accessory Protein/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Orthomyxoviridae/drug effects , Orthomyxoviridae/growth & development , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Signal Transduction , Survival Analysis , Toll-Like Receptor 2/antagonists & inhibitors , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology
18.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 23(5): 1272-6, 2015 Oct.
Article in Zh | MEDLINE | ID: mdl-26524021

ABSTRACT

OBJECTIVE: To clone the variable region genes of human anti-IL1RAP (IL-1 receptor accessory protein) monoclonal antibodies (McAb) and to construct IL1RAP chimeric antigen receptors (CARs). METHODS: The VH and VL DNA of IL1RAP single chain antibodies were amplified by RACE and overlap extension PCR from total RNA extracted from 3H6E10 and 10D8A7 hybridoma and ligated into specific IL1RAP single-chain variable fragments (scFv). CD8α transmembrane domain, CD137 intracellular domain, TCR ζ chain, human CD8α signal peptide and scFv-anti-IL1RAP were cloned into plasmid LV-lac. Recombinant lentiviruses were generated by co-transfection of recombinant plasmid LV-lac, pMD2. G, and psPAX2 helper vectors into 293FT packing cells. RESULTS: The VH and VL genes of 2 human anti-IL1RAP McAb were acquired. The 3H6E10 VH and VL genes consisted of 402 bp and 393 bp encoding 134 and 131 aminoacid residues, respectively; 10D8A7 VH and VL genes consisted of 423 bp and 381 bp encoding 141 and 127 amine acid residues, respectively. Recombinant expression vertors LV-3H6E10 scFv-ICD and LV-10D8A7 scFv-ICD (ICD: CD8α transmembrane domain-CD137 intracellular domain-TCR ζ chain) were constructed. The target fragments were demonstrated by sequencing analysis. Recombinant plasmids were transfected into 293FT cells and lentiviral particles were acquired. CONCLUSION: Human anti-IL1RAP recombinant receptors are constructed successfully and lay a good foundation for the construction of IL1RAP-CAR killer T cell vaccine.


Subject(s)
Antibodies, Monoclonal/genetics , Interleukin-1 Receptor Accessory Protein/immunology , Receptors, Antigen/genetics , Cloning, Molecular , Genetic Vectors , Humans , Hybridomas , Immunoglobulin Variable Region/genetics , Plasmids , Polymerase Chain Reaction , Single-Chain Antibodies
19.
Mol Immunol ; 63(1): 80-5, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25017307

ABSTRACT

Interleukin-33 (IL-33) is a recently discovered cytokine that belongs to the IL-1 superfamily and acts as an important regulator in several allergic disorders. It is considered to function as an alarmin, or danger cytokine, that is released upon structural cell damage. IL-33 activates several immune cells, including Th2 cells, mast cells and basophils, following its interaction with a cell surface heterodimer consisting of an IL-1 receptor-related protein ST2 (IL-1RL1) and IL-1 receptor accessory protein (IL-1RAcP). This activation leads to the production of a variety of Th2-like cytokines that mediate allergic-type immune responses. Thus, IL-33 appears to be a double-edged sword because, in addition to its important contribution to host defence, it exacerbates allergic responses, such as allergic rhinitis and asthma. A major purported mechanism of IL-33 in allergy is the activation of mast cells to produce a variety of pro-inflammatory cytokines and chemokines. In this review, we summarize the current knowledge regarding the genetics and physiology of IL-33 and IL-1RL1 and its association with different allergic diseases by focusing on its effects on mast cells and basophils.


Subject(s)
Basophils/immunology , Hypersensitivity/immunology , Interleukins/immunology , Mast Cells/immunology , Receptors, Interleukin-1 Type I/immunology , Humans , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-33 , Signal Transduction/immunology , Th2 Cells/immunology
20.
Nat Commun ; 5: 4413, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25048602

ABSTRACT

Identifying the contact regions between a protein and its binding partners is essential for creating therapies that block the interaction. Unfortunately, such contact regions are extremely difficult to characterize because they are hidden inside the binding interface. Here we introduce protein painting as a new tool that employs small molecules as molecular paints to tightly coat the surface of protein-protein complexes. The molecular paints, which block trypsin cleavage sites, are excluded from the binding interface. Following mass spectrometry, only peptides hidden in the interface emerge as positive hits, revealing the functional contact regions that are drug targets. We use protein painting to discover contact regions between the three-way interaction of IL1ß ligand, the receptor IL1RI and the accessory protein IL1RAcP. We then use this information to create peptides and monoclonal antibodies that block the interaction and abolish IL1ß cell signalling. The technology is broadly applicable to discover protein interaction drug targets.


Subject(s)
Molecular Targeted Therapy/methods , Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , Coloring Agents/chemistry , Interleukin-1 Receptor Accessory Protein/immunology , Interleukin-1 Receptor Accessory Protein/metabolism , Interleukin-1beta/metabolism , Mass Spectrometry/methods , Mice , Models, Molecular , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Protein Conformation , Receptors, Interleukin-1 Type I/metabolism , Solvents , Trypsin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL