Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Front Immunol ; 12: 699987, 2021.
Article in English | MEDLINE | ID: mdl-34552583

ABSTRACT

Objective: IL-17A plays a major role in the pathogenesis of spondyloarthritis (SpA). Here we assessed the impact of inhibition of RAR related orphan receptor-γ (RORC), the key transcription factor controlling IL-17 production, on experimental SpA in HLA-B27 transgenic (tg) rats. Methods: Experimental SpA was induced by immunization of HLA-B27 tg rats with heat-inactivated Mycobacterium tuberculosis. Splenocytes obtained at day 7, 14 and 21 after immunization were restimulated ex vivo to assess the induction of pro-inflammatory cytokines. Rats were then prophylactically treated with a RORC inhibitor versus vehicle control. The biologic effect of RORC inhibition was assessed by pro-inflammatory cytokine expression in draining lymph nodes. Arthritis and spondylitis were monitored clinically, and the degree of peripheral and axial inflammation, destruction and new bone formation was confirmed by histology. Results: Ex vivo mRNA and protein analyses revealed the rapid and selective induction of IL-17A and IL-22 production by a variety of lymphocyte subsets upon disease induction in HLA-B27 tg rats. Prophylactic RORC inhibition in vivo suppressed the expression of IL-17A, IL17F, and IL-22 without affecting the expression of other T helper cell subset related genes. This biological effect did not translate into clinical efficacy as RORC inhibition significantly accelerated the onset of arthritis and spondylitis, and aggravated the clinical severity of arthritis. This worsening of experimental SpA was confirmed by histopathological demonstration of increased inflammation, destruction, and new bone formation. Conclusion: Despite a significant suppression of the IL-17 axis, RORC inhibitor treatment accelerates and aggravates experimental SpA in the HLA-B27 tg rat model.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Spondylarthritis/immunology , Spondylarthritis/pathology , Animals , Disease Models, Animal , Female , HLA-B27 Antigen/genetics , Male , Rats , Rats, Inbred Lew , Rats, Transgenic
2.
Nat Commun ; 10(1): 9, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30602780

ABSTRACT

Dysregulated IL-23/IL-17 responses have been linked to psoriatic arthritis and other forms of spondyloarthritides (SpA). RORγt, the key Thelper17 (Th17) cell transcriptional regulator, is also expressed by subsets of innate-like T cells, including invariant natural killer T (iNKT) and γδ-T cells, but their contribution to SpA is still unclear. Here we describe the presence of particular RORγt+T-betloPLZF- iNKT and γδ-hi T cell subsets in healthy peripheral blood. RORγt+ iNKT and γδ-hi T cells show IL-23 mediated Th17-like immune responses and were clearly enriched within inflamed joints of SpA patients where they act as major IL-17 secretors. SpA derived iNKT and γδ-T cells showed unique and Th17-skewed phenotype and gene expression profiles. Strikingly, RORγt inhibition blocked γδ17 and iNKT17 cell function while selectively sparing IL-22+ subsets. Overall, our findings highlight a unique diversity of human RORγt+ T cells and underscore the potential of RORγt antagonism to modulate aberrant type 17 responses.


Subject(s)
Natural Killer T-Cells/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Spondylarthritis/immunology , T-Lymphocyte Subsets/metabolism , Case-Control Studies , Humans , Interleukin-17/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Receptors, Interleukin/metabolism
3.
J Biol Chem ; 292(28): 11618-11630, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28546429

ABSTRACT

The nuclear receptor retinoid acid receptor-related orphan receptor γt (RORγt) is a master regulator of the Th17/IL-17 pathway that plays crucial roles in the pathogenesis of autoimmunity. RORγt has recently emerged as a highly promising target for treatment of a number of autoimmune diseases. Through high-throughput screening, we previously identified several classes of inverse agonists for RORγt. Here, we report the crystal structures for the ligand-binding domain of RORγt in both apo and ligand-bound states. We show that apo RORγt adopts an active conformation capable of recruiting coactivator peptides and present a detailed analysis of the structural determinants that stabilize helix 12 (H12) of RORγt in the active state in the absence of a ligand. The structures of ligand-bound RORγt reveal that binding of the inverse agonists disrupts critical interactions that stabilize H12. This destabilizing effect is supported by ab initio calculations and experimentally by a normalized crystallographic B-factor analysis. Of note, the H12 destabilization in the active state shifts the conformational equilibrium of RORγt toward an inactive state, which underlies the molecular mechanism of action for the inverse agonists reported here. Our findings highlight that nuclear receptor structure and function are dictated by a dynamic conformational equilibrium and that subtle changes in ligand structures can shift this equilibrium in opposite directions, leading to a functional switch from agonists to inverse agonists.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Inverse Agonism , Models, Molecular , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Apoproteins/antagonists & inhibitors , Apoproteins/chemistry , Apoproteins/genetics , Apoproteins/metabolism , Binding Sites , Binding, Competitive , Cells, Cultured , Genes, Reporter/drug effects , HEK293 Cells , Humans , Interleukin-17/antagonists & inhibitors , Interleukin-17/metabolism , Ligands , Molecular Conformation , Nuclear Receptor Subfamily 1, Group F, Member 3/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylalanine/metabolism , Phenylalanine/pharmacology , Phenylurea Compounds/chemistry , Phenylurea Compounds/metabolism , Phenylurea Compounds/pharmacology , Protein Conformation , Protein Interaction Domains and Motifs , Protein Refolding , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism
4.
Eur J Immunol ; 38(5): 1204-14, 2008 May.
Article in English | MEDLINE | ID: mdl-18412165

ABSTRACT

Epstein-Barr virus-induced gene 3 (EBI3) associates with p28 to form IL-27 and with IL-12p35 to form IL-35. IL-27Ralpha(-/-) mice studies indicate that IL-27 negatively regulates Th17 cell differentiation. However, no EBI3, p28 or p35-deficiency studies that directly address the role of EBI3, p28 or p35 on Th17 cells have been done. Here, we demonstrate that spleen cells derived from EBI3(-/-) mice produce significantly higher levels of IL-17 as well as IL-22 upon stimulation with OVA. In vitro derived EBI3(-/-) Th17 cells also produced significantly higher levels of IL-17 and IL-22 than WT cells. The frequency of IL-17-producing cells was also elevated when EBI3(-/-) cells were cultured under Th17 conditions. In addition, spleen cells from EBI3(-/-) mice immunized with Listeria monocytogenes produced significantly elevated levels of IL-17 and IL-22. Furthermore, the Th17 transcription factor RORgamma t was significantly enhanced in EBI3(-/-) cells. Finally, EBI3(-/-) mice exhibited a reduced bacterial load following an acute challenge with L. monocytogenes or a re-challenge of previously immunized mice, suggesting that EBI3 negatively regulates both innate and adaptive immunity. Taken together, these data provide direct evidence that EBI3 negatively regulates the expression of IL-17, IL-22 and RORgamma t as well as protective immunity against L. monocytogenes.


Subject(s)
Gene Expression Regulation , Interleukin-17/metabolism , Interleukins/genetics , Interleukins/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Thyroid Hormone/genetics , T-Lymphocytes/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression/genetics , Interferon-gamma/blood , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-17/blood , Interleukin-17/genetics , Listeria monocytogenes/immunology , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Minor Histocompatibility Antigens , Nuclear Receptor Subfamily 1, Group F, Member 3 , Ovalbumin/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Spleen/cytology , Spleen/microbiology , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Interleukin-22
5.
Biochem Pharmacol ; 74(1): 98-106, 2007 Jun 30.
Article in English | MEDLINE | ID: mdl-17482579

ABSTRACT

Molecular K(d) and k(off) parameters are often used to define the molecular potency of drugs. These constants, however, are determined on purified target proteins, and their relationship to in vivo binding phenomena is poorly understood. Herein, we report two novel assays to determine the off-rates of allosteric antagonists from lymphocyte function-associated antigen 1 (LFA-1). The SPR assay involves using the non-blocking mAb TS2/4 to immobilize full-length LFA-1 on a hydrophilic chip surface, and the soluble, native ligand sICAM-1 to probe the fraction of free LFA-1. To determine the fraction of free LFA-1 on cell surfaces, a flow cytometry assay was developed utilizing the fluorophore-labeled Fab R3.1. The R3.1 antibody has been previously demonstrated to block the ability of both ICAM-1 and antagonists to bind to purified and cell-surface LFA-1. The molecular and ex vivo cellular parameters were determined for a set of nine structurally-related LFA-1 allosteric antagonists. The relationships between the parameters determined in the ELISA (K(d)), SPR (k(off)), and flow cytometry (k(off)) assays were shown to be linear with slopes approximately equal to 1, and a correlation analysis showed that the three assay datasets were equivalent at the alpha=0.05 level. These results were unexpected, as the ELISA and SPR assays involve high affinity LFA-1, and the flow cytometry assays involve cell surface LFA-1 in whole-blood, in which a distribution of affinity states would be expected. Nevertheless, the results presented herein show that the K(d) and k(off)'s determined in molecular assays can be used as predictors of LFA-1 receptor occupancy in ex vivo assays.


Subject(s)
Cell Adhesion Molecules/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Imidazolidines/metabolism , Integrins/antagonists & inhibitors , Lymphocyte Function-Associated Antigen-1/metabolism , Surface Plasmon Resonance/methods , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Allosteric Site/drug effects , Allosteric Site/physiology , Antibodies, Monoclonal/metabolism , Cell Adhesion Molecules/chemistry , Flow Cytometry , Hydantoins/chemistry , Hydantoins/metabolism , Hydantoins/pharmacology , Imidazolidines/chemistry , Imidazolidines/pharmacology , Integrins/immunology , Intercellular Adhesion Molecule-1/pharmacology , Kinetics , Lymphocyte Function-Associated Antigen-1/chemistry , Reproducibility of Results
6.
Biophys Chem ; 128(2-3): 156-64, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17466438

ABSTRACT

The glucocorticoid receptor (GR) is involved in the transcriptional regulation of genes associated with inflammation, glucose homeostasis, and bone turnover through the association with ligands, such as corticosteroids. GR-mediated gene transcription is regulated or fine-tuned via the recruitment of co-factors including coactivators and corepressors. Current therapeutic approaches to targeting GR aim to retain the beneficial anti-inflammatory activity of the corticosteroids while eliminating negative side effects. Towards achieving this goal the experiments discussed here reveal a mechanism of co-factor binding in the presence of either bound agonist or antagonist. The GR ligand binding domain (GR-LBD(F602S)), in the presence of agonist or antagonist, utilizes different modes of binding for coactivator versus corepressor. Coactivator binding to the co-effector binding pocket of GR-LBD(F602S) is driven both by favorable enthalpic and entropic interactions whereas corepressor binding to the same pocket is entropically driven. These data support the hypothesis that ligand-induced conformational changes dictate co-factor binding and subsequent trans-activation or trans-repression.


Subject(s)
Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Amino Acid Sequence , Circular Dichroism , Dexamethasone/chemistry , Kinetics , Ligands , Mifepristone/chemistry , Peptides/chemistry , Protein Binding , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Thermodynamics
7.
Bioorg Med Chem Lett ; 17(9): 2465-9, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17379516

ABSTRACT

The synthesis and in vitro activities of a series of succinyl-nitrile-based inhibitors of Cathepsin S are described. Several members of this class show nanomolar inhibition of the target enzyme as well as cellular potency. The inhibitors displaying the greatest potency contain N-alkyl substituted piperidine and pyrrolidine rings spiro-fused to the alpha-carbon of the P1 residue.


Subject(s)
Cathepsins/antagonists & inhibitors , Chemistry, Pharmaceutical/methods , Nitriles/chemistry , Catalytic Domain , Dipeptides/chemistry , Drug Design , Humans , Models, Chemical , Molecular Conformation , Nitriles/classification , Peptides/chemistry , Piperidines/chemistry , Pyrrolidines/chemistry , Structure-Activity Relationship
8.
Eur J Immunol ; 36(12): 3305-14, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17111354

ABSTRACT

Protein kinase C theta (PKCtheta) is essential for T cell activation, as it is required for the activation of NF-kappaB and expression of IL-2. PKCtheta has also been shown to affect NFAT activation and Th2 differentiation. To better understand the role of PKCtheta in the regulation of T helper cells, we used PKCtheta-deficient DO11.10 transgenic T cells to study its role in vitro. DO11.10 Th1 cells deficient in PKCtheta produced significantly less TNF-alpha and IL-2. The expression of Th2 cytokines, including IL-4, IL-5, IL-10, IL-13 and IL-24 was significantly reduced in PKCtheta-deficient T cells. Moreover, the expression of the Th2 transcription factor, GATA3, was significantly reduced in PKCtheta-deficient T cells. Overexpression of GATA3 by retroviral infection in PKCtheta-deficient T cells resulted in increased expansion of IL-4-producing T cells and higher IL-4 production than that of wild type Th2 cells. IL-5, IL-10, IL-13 and IL-24 expressions were also rescued by GATA3 overexpression. Our observations suggest that PKCtheta regulates Th2 cytokine expression via GATA3.


Subject(s)
Cytokines/biosynthesis , GATA3 Transcription Factor/physiology , Isoenzymes/physiology , Protein Kinase C/physiology , Th2 Cells/enzymology , Animals , Cells, Cultured , Cytokines/genetics , GATA3 Transcription Factor/biosynthesis , GATA3 Transcription Factor/genetics , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Protein Kinase C-theta , Th2 Cells/immunology , Th2 Cells/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
9.
Biochemistry ; 43(37): 11658-71, 2004 Sep 21.
Article in English | MEDLINE | ID: mdl-15362850

ABSTRACT

A novel inhibitor of p38 mitogen-activated protein kinase (p38), CMPD1, identified by high-throughput screening, is characterized herein. Unlike the p38 inhibitors described previously, this inhibitor is substrate selective and noncompetitive with ATP. In steady-state kinetics experiments, CMPD1 was observed to prevent the p38alpha-dependent phosphorylation (K(i)(app) = 330 nM) of the splice variant of mitogen-activated protein kinase-activated protein kinase 2 (MK2a) that contains a docking domain for p38alpha and p38beta, but it did not prevent the phosphorylation of ATF-2 (K(i)(app) > 20 microM). In addition to kinetic studies, isothermal titration calorimetry and surface plasmon resonance experiments were performed to elucidate the mechanism of inhibition. While isothermal titration calorimetry analysis indicated that CMPD1 binds to p38alpha, CMPD1 was not observed to compete with ATP for p38alpha, nor was it able to interrupt the binding of p38alpha to MK2a observed by surface plasmon resonance. Therefore, deuterium exchange mass spectrometry (DXMS) was employed to study the p38alpha.CMPD1 inhibitory complex, to provide new insight into the mechanism of substrate selective inhibition. The DXMS data obtained for the p38alpha.CMPD1 complex were compared to the data obtained for the p38alpha.MK2a complex and a p38alpha.active site binding inhibitor complex. Alterations in the DXMS behavior of both p38alpha and MK2a were observed upon complex formation, including but not limited to the interaction between the carboxy-terminal docking domain of MK2a and its binding groove on p38alpha. Alterations in the D(2)O exchange of p38alpha produced by CMPD1 suggest that the substrate selective inhibitor binds in the vicinity of the active site of p38alpha, resulting in perturbations to regions containing nucleotide binding pocket residues, docking groove residues (E160 and D161), and a Mg(2+) ion cofactor binding residue (D168). Although the exact mechanism of substrate selective inhibition by this novel inhibitor has not yet been disclosed, the results suggest that CMPD1 binding in the active site region of p38alpha induces perturbations that may result in the suboptimal positioning of substrates and cofactors in the transition state, resulting in selective inhibition of p38alpha activity.


Subject(s)
Biphenyl Compounds/metabolism , Enzyme Inhibitors/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Structure, Tertiary , Activating Transcription Factor 2 , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Binding Sites , Biphenyl Compounds/chemistry , Calorimetry , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Inhibitors/chemistry , Intracellular Signaling Peptides and Proteins , Mice , Mitogen-Activated Protein Kinase 14 , Mitogen-Activated Protein Kinases/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Phosphorylation , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Substrate Specificity , Surface Plasmon Resonance , Transcription Factors/metabolism
10.
Biochemistry ; 43(31): 9950-60, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15287722

ABSTRACT

The p38 mitogen-activated protein kinase (p38) pathway is required for the production of proinflammatory cytokines (TNFalpha and IL-1) that mediate the chronic inflammatory phases of several autoimmune diseases. Potent p38 inhibitors, such as the slow tight-binding inhibitor BIRB 796, have recently been reported to block the production of TNFalpha and IL-1beta. Here we analyze downstream signaling complexes and molecular mechanisms, to provide new insight into the function of p38 signaling complexes and the development of novel inhibitors of the p38 pathway. Catalysis, signaling functions, and molecular interactions involving p38alpha and one of its downstream signaling partners, mitogen-activated protein kinase-activated protein kinase 2 (MK2), have been explored by steady-state kinetics, surface plasmon resonance, isothermal calorimetry, and stopped-flow fluorescence. Functional 1/1 signaling complexes (Kd = 1-100 nM) composed of activated and nonactivated forms of p38alpha and a splice variant of MK2 (MK2a) were characterized. Catalysis of MK2a phosphorylation and activation by p38alpha was observed to be efficient under conditions where substrate is saturating (kcat(app) = 0.05-0.3 s(-1)) and nonsaturating (kcat(app)/KM(app) = 1-3 x 10(6) M(-1) s(-1)). Specific interactions between the carboxy-terminal residues of MK2a (370-400) and p38alpha precipitate formation of a high-affinity complex (Kd = 20 nM); the p38alpha-dependent MK2a phosphorylation reaction was inhibited by the 30-amino acid docking domain peptide of MK2a (IC50 = 60 nM). The results indicate that the 30-amino acid docking domain peptide of MK2a is required for the formation of a tight, functional p38alpha.MK2a complex, and that perturbation of the tight-docking interaction between these signaling partners prevents the phosphorylation of MK2a. The thermodynamic and steady-state kinetic characterization of the p38alpha.MK2a signaling complex has led to a clear understanding of complex formation, catalysis, and function on the molecular level.


Subject(s)
MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/chemistry , Mitogen-Activated Protein Kinases/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology , Alternative Splicing , Animals , Calorimetry , Catalysis , Fluorescein-5-isothiocyanate/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/physiology , Kinetics , MAP Kinase Signaling System/genetics , Mice , Mitogen-Activated Protein Kinase 14 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/genetics , Peptide Fragments/physiology , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary/genetics , Spectrometry, Fluorescence , Structure-Activity Relationship , Surface Plasmon Resonance , Thermodynamics , Ultracentrifugation
11.
Bioorg Med Chem ; 11(5): 733-40, 2003 Mar 06.
Article in English | MEDLINE | ID: mdl-12538003

ABSTRACT

The design and synthesis of dipeptidyl disulfides and dipeptidyl benzoylhydrazones as selective inhibitors of the cysteine protease Cathepsin S are described. These inhibitors were expected to form a slowly reversible covalent adduct of the active site cysteine of Cathepsin S. Formation of the initial adduct was confirmed by mass spectral analysis. The nature and mechanism of these adducts was explored. Kinetic analysis of the benzoyl hydrazones indicate that these inhibitors are acting as irreversible inhibitors of Cathepsin S. Additionally, the benzoylhydrazones were shown to be potent inhibitors of Cathepsin S processing of Class II associated invariant peptide both in vitro and in vivo.


Subject(s)
Cathepsins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemical synthesis , Cysteine Proteinase Inhibitors/pharmacology , Disulfides/chemical synthesis , Disulfides/pharmacology , Hydrazones/chemical synthesis , Hydrazones/pharmacology , Animals , Cathepsin B/antagonists & inhibitors , Cell Line , Drug Design , Humans , Kinetics , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pancreatic Elastase/antagonists & inhibitors , Precipitin Tests , Recombinant Proteins/antagonists & inhibitors
12.
J Med Chem ; 45(25): 5471-82, 2002 Dec 05.
Article in English | MEDLINE | ID: mdl-12459015

ABSTRACT

The specificity of the immune response relies on processing of foreign proteins and presentation of antigenic peptides at the cell surface. Inhibition of antigen presentation, and the subsequent activation of T-cells, should, in theory, modulate the immune response. The cysteine protease Cathepsin S performs a fundamental step in antigen presentation and therefore represents an attractive target for inhibition. Herein, we report a series of potent and reversible Cathepsin S inhibitors based on dipeptide nitriles. These inhibitors show nanomolar inhibition of the target enzyme as well as cellular potency in a human B cell line. The first X-ray crystal structure of a reversible inhibitor cocrystallized with Cathepsin S is also reported.


Subject(s)
Cathepsins/chemical synthesis , Dipeptides/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Nitriles/chemical synthesis , B-Lymphocytes/drug effects , Binding, Competitive , Cathepsins/chemistry , Cathepsins/pharmacology , Cell Line , Crystallography, X-Ray , Dipeptides/chemistry , Dipeptides/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Models, Molecular , Nitriles/chemistry , Nitriles/pharmacology , Stereoisomerism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...