Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
ACS Pharmacol Transl Sci ; 6(9): 1275-1287, 2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37705593

ABSTRACT

Hematopoietic cell transplantation (HCT) is a proven and potentially curable therapy for hematological malignancies and inherited hematological disease. The main risk of HCT is the development of graft versus host disease (GVHD) acquired in up to 50% of patients. Upregulation of soluble ST2 (sST2) is a key clinical biomarker for GVHD prognosis and was shown to be a potential therapeutic target for GVHD. Agents targeting sST2 to reduce the sST2 level after HCT have the potential to mitigate GVHD progression. Here, we report 32 (or XY52) as the lead ST2 inhibitor from our optimization campaign. XY52 had improved inhibitory activity and metabolic stability in vitro and in vivo. XY52 suppressed proinflammatory T-cell proliferation while increasing regulatory T cells in vitro. In a clinically relevant GVHD model, a 21-day prophylactic regimen of XY52 reduced plasma sST2 and IFN-γ levels and GVHD score and extended survival in mice. XY52 represented a significant improvement over our previous compound, iST2-1, and further optimization of XY52 is warranted. The small-molecule ST2 inhibitors can potentially be used as a biomarker-guided therapy for mitigating GVHD in future clinical applications.

2.
Bioorg Med Chem ; 71: 116942, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35930851

ABSTRACT

An elevated plasma level of soluble ST2 (sST2) is a risk biomarker for graft-versus-host disease (GVHD) and death in patients receiving hematopoietic cell transplantation (HCT). sST2 functions as a trap for IL-33 and amplifies the pro-inflammatory type 1 and 17 response while suppressing the tolerogenic type 2 and regulatory T cells activation during GVHD development. We previously identified small-molecule ST2 inhibitors particularly iST2-1 that reduces plasma sST2 levels and improved survival in two animal models. Here, we reported the structure-activity relationship of the furanylmethylpyrrolidine-based ST2 inhibitors based on iST2-1. Based on the biochemical AlphaLISA assay, we improved the activity of iST2-1 by 6-fold (∼6 µM in IC50 values) in the inhibition of ST2/IL-33 and confirmed the activities of the compounds in a cellular reporter assay. To determine the inhibition of the alloreactivity in vitro, we used the mixed lymphocyte reaction assay to demonstrate that our ST2 inhibitors decreased CD4+ and CD8+ T cells proliferation and increased Treg population. The data presented in this work are critical to the development of ST2 inhibitors in future.


Subject(s)
Graft vs Host Disease , Animals , CD8-Positive T-Lymphocytes/metabolism , Furans , Graft vs Host Disease/drug therapy , Graft vs Host Disease/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Pyrrolidines/pharmacology , Structure-Activity Relationship
3.
J Clin Invest ; 132(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35439173

ABSTRACT

CD13, an ectoenzyme on myeloid and stromal cells, also circulates as a shed, soluble protein (sCD13) with powerful chemoattractant, angiogenic, and arthritogenic properties, which require engagement of a G protein-coupled receptor (GPCR). Here we identify the GPCR that mediates sCD13 arthritogenic actions as the bradykinin receptor B1 (B1R). Immunofluorescence and immunoblotting verified high expression of B1R in rheumatoid arthritis (RA) synovial tissue and fibroblast-like synoviocytes (FLSs), and demonstrated binding of sCD13 to B1R. Chemotaxis, and phosphorylation of Erk1/2, induced by sCD13, were inhibited by B1R antagonists. In ex vivo RA synovial tissue organ cultures, a B1R antagonist reduced secretion of inflammatory cytokines. Several mouse arthritis models, including serum transfer, antigen-induced, and local innate immune stimulation arthritis models, were attenuated in Cd13-/- and B1R-/- mice and were alleviated by B1R antagonism. These results establish a CD13/B1R axis in the pathogenesis of inflammatory arthritis and identify B1R as a compelling therapeutic target in RA and potentially other inflammatory diseases.


Subject(s)
Arthritis, Rheumatoid , CD13 Antigens/metabolism , Synoviocytes , Animals , Arthritis, Rheumatoid/pathology , Bradykinin/metabolism , Bradykinin/pharmacology , Disease Models, Animal , Fibroblasts/metabolism , Mice , Receptor, Bradykinin B1/genetics , Receptor, Bradykinin B1/metabolism , Receptors, G-Protein-Coupled/metabolism , Synovial Membrane/pathology , Synoviocytes/metabolism
4.
Cancer Res Commun ; 2(12): 1727-1737, 2022 12.
Article in English | MEDLINE | ID: mdl-36970721

ABSTRACT

The reciprocal relationship between malignant T cells and lymphoma-associated macrophages (LAM) within the tumor microenvironment (TME) is unique, as LAMs are well poised to provide ligands for antigen, costimulatory, and cytokine receptors that promote T-cell lymphoma growth. Conversely, malignant T cells promote the functional polarization and homeostatic survival of LAM. Therefore, we sought to determine the extent to which LAMs are a therapeutic vulnerability in these lymphomas, and to identify effective therapeutic strategies for their depletion. We utilized complementary genetically engineered mouse models and primary peripheral T-cell lymphoma (PTCL) specimens to quantify LAM expansion and proliferation. A high-throughput screen was performed to identify targeted agents that effectively deplete LAM within the context of PTCL. We observed that LAMs are dominant constituents of the TME in PTCL. Furthermore, their dominance was explained, at least in part, by their proliferation and expansion in response to PTCL-derived cytokines. Importantly, LAMs are a true dependency in these lymphomas, as their depletion significantly impaired PTCL progression. These findings were extrapolated to a large cohort of human PTCL specimens where LAM proliferation was observed. A high-throughput screen demonstrated that PTCL-derived cytokines led to relative resistance to CSF1R selective inhibitors, and culminated in the identification of dual CSF1R/JAK inhibition as a novel therapeutic strategy to deplete LAM in these aggressive lymphomas. Malignant T cells promote the expansion and proliferation of LAM, which are a bone fide dependency in these lymphomas, and are effectively depleted with a dual CSF1R/JAK inhibitor. Significance: LAMs are a therapeutic vulnerability, as their depletion impairs T-cell lymphoma disease progression. Pacritinib, a dual CSF1R/JAK inhibitor, effectively impaired LAM viability and expansion, prolonged survival in preclinical T-cell lymphoma models, and is currently being investigated as a novel therapeutic approach in these lymphomas.


Subject(s)
Janus Kinase Inhibitors , Lymphoma, T-Cell, Peripheral , Lymphoma, T-Cell , Animals , Mice , Humans , Lymphoma, T-Cell, Peripheral/drug therapy , Janus Kinase Inhibitors/pharmacology , Cytokines/pharmacology , Lymphoma, T-Cell/drug therapy , Macrophages , Tumor Microenvironment
5.
JCI Insight ; 3(14)2018 07 26.
Article in English | MEDLINE | ID: mdl-30046004

ABSTRACT

Soluble cytokine receptors function as decoy receptors to attenuate cytokine-mediated signaling and modulate downstream cellular responses. Dysregulated overproduction of soluble receptors can be pathological, such as soluble ST2 (sST2), a prognostic biomarker in cardiovascular diseases, ulcerative colitis, and graft-versus-host disease (GVHD). Although intervention using an ST2 antibody improves survival in murine GVHD models, sST2 is a challenging target for drug development because it binds to IL-33 via an extensive interaction interface. Here, we report the discovery of small-molecule ST2 inhibitors through a combination of high-throughput screening and computational analysis. After in vitro and in vivo toxicity assessment, 3 compounds were selected for evaluation in 2 experimental GVHD models. We show that the most effective compound, iST2-1, reduces plasma sST2 levels, alleviates disease symptoms, improves survival, and maintains graft-versus-leukemia activity. Our data suggest that iST2-1 warrants further optimization to develop treatment for inflammatory diseases mediated by sST2.


Subject(s)
Drug Discovery , Interleukin-1 Receptor-Like 1 Protein/drug effects , Interleukin-1 Receptor-Like 1 Protein/metabolism , Proteomics , Receptors, Cytokine/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biomarkers , Cell Line, Tumor , Computational Biology , Drug Evaluation, Preclinical , Graft vs Host Disease , High-Throughput Screening Assays , Interleukin-33/metabolism , Leukemia/drug therapy , Mice , Models, Animal , Stem Cell Transplantation
6.
Lupus Sci Med ; 3(1): e000147, 2016.
Article in English | MEDLINE | ID: mdl-27099767

ABSTRACT

OBJECTIVES: Antigen-specific CD4(+) T cells epigenetically modified with DNA methylation inhibitors overexpress genes normally suppressed by this mechanism, including CD11a, CD70, CD40L and the KIR gene family. The altered cells become autoreactive, losing restriction for nominal antigen and responding to self-class II major histocompatibility complex (MHC) molecules without added antigen, and are sufficient to cause a lupus-like disease in syngeneic mice. T cells overexpressing the same genes are found in patients with active lupus. Whether these genes are co-overexpressed on the same or different cells is unknown. The goal of this study was to determine whether these genes are overexpressed on the same or different T cells and whether this subset of CD4(+) T cells is also present in patients with lupus and other rheumatic diseases. METHODS: Multicolour flow cytometry was used to compare CD11a, CD70, CD40L and KIR expression on CD3(+)CD4(+)CD28(+) T cells to their expression on experimentally demethylated CD3(+)CD4(+)CD28(+) T cells and CD3(+)CD4(+)CD28(+) T cells from patients with active lupus and other autoimmune diseases. RESULTS: Experimentally demethylated CD4(+) T cells and T cells from patients with active lupus have a CD3(+)CD4(+)CD28(+)CD11a(hi)CD70(+)CD40L(hi)KIR(+) subset, and the subset size is proportional to lupus flare severity. A similar subset is found in patients with other rheumatic diseases including rheumatoid arthritis, systemic sclerosis and Sjögren's syndrome but not retroperitoneal fibrosis. CONCLUSIONS: Patients with active autoimmune rheumatic diseases have a previously undescribed CD3(+)CD4(+)CD28(+)CD11a(hi)CD70(+)CD40L(hi)KIR(+) T cell subset. This subset may play an important role in flares of lupus and related autoimmune rheumatic diseases, provide a biomarker for disease activity and serve as a novel therapeutic target for the treatment of lupus flares.

7.
J Mol Biol ; 394(3): 391-409, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19733184

ABSTRACT

Bimolecular fluorescence complementation (BiFC) analysis enables visualization of the subcellular locations of protein interactions in living cells. Using fragments of different fluorescent proteins, we investigated the temporal resolution and the quantitative accuracy of BiFC analysis. We determined the kinetics of BiFC complex formation in response to the rapamycin-inducible interaction between the FK506 binding protein (FKBP) and the FKBP-rapamycin binding domain (FRB). Fragments of yellow fluorescent protein fused to FKBP and FRB produced detectable BiFC complex fluorescence 10 min after the addition of rapamycin and a 10-fold increase in the mean fluorescence intensity in 8 h. The N-terminal fragment of the Venus fluorescent protein fused to FKBP produced constitutive BiFC complexes with several C-terminal fragments fused to FRB. A chimeric N-terminal fragment containing residues from Venus and yellow fluorescent protein produced either constitutive or inducible BiFC complexes depending on the temperature at which the cells were cultured. The concentrations of inducers required for half-maximal induction of BiFC complex formation by all fluorescent protein fragments tested were consistent with the affinities of the inducers for unmodified FKBP and FRB. Treatment with the FK506 inhibitor of FKBP-FRB interaction prevented the formation of BiFC complexes by FKBP and FRB fusions, but did not disrupt existing BiFC complexes. Proteins synthesized before the addition of rapamycin formed BiFC complexes with the same efficiency as did newly synthesized proteins. Inhibitors of protein synthesis attenuated BiFC complex formation independent of their effects on fusion protein synthesis. The kinetics at which they inhibited BiFC complex formation suggests that they prevented association of the fluorescent protein fragments, but not the slow maturation of BiFC complex fluorescence. Agents that induce the unfolded protein response also reduced formation of BiFC complexes. The effects of these agents were suppressed by cellular adaptation to protein folding stress. In summary, BiFC analysis enables detection of protein interactions within minutes after complex formation in living cells, but does not allow detection of complex dissociation. Conditional BiFC complex formation depends on the folding efficiencies of fluorescent protein fragments and can be affected by the cellular protein folding environment.


Subject(s)
Luminescent Proteins/chemistry , Animals , COS Cells , Chlorocebus aethiops , Cycloheximide , Flow Cytometry , In Vitro Techniques , Kinetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Models, Biological , Multiprotein Complexes/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Folding/drug effects , Protein Interaction Domains and Motifs , Protein Synthesis Inhibitors/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Signal Transduction , Tacrolimus/pharmacology , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/metabolism
8.
FEBS Lett ; 555(2): 236-42, 2003 Dec 04.
Article in English | MEDLINE | ID: mdl-14644421

ABSTRACT

Cytochrome p450 (CYP) 4Fs metabolize leukotriene B(4) and other inflammatory mediators in the arachidonic acid cascade. Here we show that lipopolysaccharide (LPS) treatment suppresses CYP4F4 and up-regulates CYP4F5 mRNA expression in rat liver whereas renal CYP4Fs are essentially unchanged. BaSO(4) treatment, in contrast, increases both hepatic and renal CYP4F expression levels. Thus, distinct regulatory mechanisms in CYP4F expression might operate under different inflammatory prompts. To examine hepatic totipotency, primary hepatocytes were treated with varying doses of LPS resulting in decrease in all the CYP4F isoforms. Treatment of hepatocytes with 5 ng/ml of interleukin-1beta mimics the in vivo effects of LPS on CYP4F expression.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Kidney/enzymology , Lipopolysaccharides/pharmacology , Microsomes, Liver/enzymology , Mixed Function Oxygenases/metabolism , Animals , Barium Sulfate/pharmacology , Cytochrome P-450 Enzyme System/biosynthesis , Cytochrome P450 Family 4 , Gene Expression Regulation, Enzymologic , Hepatocytes/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-1/pharmacology , Isoenzymes , Male , Mixed Function Oxygenases/biosynthesis , Polymerase Chain Reaction/methods , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...