Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cell ; 186(14): 2995-3012.e15, 2023 07 06.
Article in English | MEDLINE | ID: mdl-37321220

ABSTRACT

Wnt ligands oligomerize Frizzled (Fzd) and Lrp5/6 receptors to control the specification and activity of stem cells in many species. How Wnt signaling is selectively activated in different stem cell populations, often within the same organ, is not understood. In lung alveoli, we show that distinct Wnt receptors are expressed by epithelial (Fzd5/6), endothelial (Fzd4), and stromal (Fzd1) cells. Fzd5 is uniquely required for alveolar epithelial stem cell activity, whereas fibroblasts utilize distinct Fzd receptors. Using an expanded repertoire of Fzd-Lrp agonists, we could activate canonical Wnt signaling in alveolar epithelial stem cells via either Fzd5 or, unexpectedly, non-canonical Fzd6. A Fzd5 agonist (Fzd5ag) or Fzd6ag stimulated alveolar epithelial stem cell activity and promoted survival in mice after lung injury, but only Fzd6ag promoted an alveolar fate in airway-derived progenitors. Therefore, we identify a potential strategy for promoting regeneration without exacerbating fibrosis during lung injury.


Subject(s)
Lung Injury , Mice , Animals , Wnt Proteins , Frizzled Receptors , Wnt Signaling Pathway , Alveolar Epithelial Cells , Stem Cells
2.
Nat Immunol ; 23(4): 568-580, 2022 04.
Article in English | MEDLINE | ID: mdl-35314846

ABSTRACT

Tumor-associated macrophages are composed of distinct populations arising from monocytes or tissue macrophages, with a poorly understood link to disease pathogenesis. Here, we demonstrate that mouse monocyte migration was supported by glutaminyl-peptide cyclotransferase-like (QPCTL), an intracellular enzyme that mediates N-terminal modification of several substrates, including the monocyte chemoattractants CCL2 and CCL7, protecting them from proteolytic inactivation. Knockout of Qpctl disrupted monocyte homeostasis, attenuated tumor growth and reshaped myeloid cell infiltration, with loss of monocyte-derived populations with immunosuppressive and pro-angiogenic profiles. Antibody targeting of the receptor CSF1R, which more broadly eliminates tumor-associated macrophages, reversed tumor growth inhibition in Qpctl-/- mice and prevented lymphocyte infiltration. Modulation of QPCTL synergized with anti-PD-L1 to expand CD8+ T cells and limit tumor growth. QPCTL inhibition constitutes an effective approach for myeloid cell-targeted cancer immunotherapy.


Subject(s)
Aminoacyltransferases , CD8-Positive T-Lymphocytes , Chemokines , Neoplasms , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Animals , CD8-Positive T-Lymphocytes/pathology , Chemokines/metabolism , Immunotherapy , Leukemic Infiltration , Mice , Mice, Knockout , Monocytes , Neoplasms/immunology
3.
Nat Immunol ; 20(4): 471-481, 2019 04.
Article in English | MEDLINE | ID: mdl-30778241

ABSTRACT

Foxp3+ regulatory T cells (Treg cells) are crucial for the maintenance of immune homeostasis both in lymphoid tissues and in non-lymphoid tissues. Here we demonstrate that the ability of intestinal Treg cells to constrain microbiota-dependent interleukin (IL)-17-producing helper T cell (TH17 cell) and immunoglobulin A responses critically required expression of the transcription factor c-Maf. The terminal differentiation and function of several intestinal Treg cell populations, including RORγt+ Treg cells and follicular regulatory T cells, were c-Maf dependent. c-Maf controlled Treg cell-derived IL-10 production and prevented excessive signaling via the kinases PI(3)K (phosphatidylinositol-3-OH kinase) and Akt and the metabolic checkpoint kinase complex mTORC1 (mammalian target of rapamycin) and expression of inflammatory cytokines in intestinal Treg cells. c-Maf deficiency in Treg cells led to profound dysbiosis of the intestinal microbiota, which when transferred to germ-free mice was sufficient to induce exacerbated intestinal TH17 responses, even in a c-Maf-competent environment. Thus, c-Maf acts to preserve the identity and function of intestinal Treg cells, which is essential for the establishment of host-microbe symbiosis.


Subject(s)
Immunoglobulin A/biosynthesis , Intestines/immunology , Microbiota , Proto-Oncogene Proteins c-maf/physiology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Cells, Cultured , Colitis/immunology , Cytokines/metabolism , Dysbiosis , Gene Expression Regulation , Homeostasis , Interleukin-10/biosynthesis , Mice, Inbred C57BL , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/metabolism , T-Lymphocytes, Regulatory/enzymology
5.
Mol Cell ; 69(4): 551-565.e7, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29452636

ABSTRACT

Inflammatory responses mediated by NOD2 rely on RIP2 kinase and ubiquitin ligase XIAP for the activation of nuclear factor κB (NF-κB), mitogen-activated protein kinases (MAPKs), and cytokine production. Herein, we demonstrate that selective XIAP antagonism blocks NOD2-mediated inflammatory signaling and cytokine production by interfering with XIAP-RIP2 binding, which removes XIAP from its ubiquitination substrate RIP2. We also establish that the kinase activity of RIP2 is dispensable for NOD2 signaling. Rather, the conformation of the RIP2 kinase domain functions to regulate binding to the XIAP-BIR2 domain. Effective RIP2 kinase inhibitors block NOD2 signaling by disrupting RIP2-XIAP interaction. Finally, we identify NOD2 signaling and XIAP-dependent ubiquitination sites on RIP2 and show that mutating these lysine residues adversely affects NOD2 pathway signaling. Overall, these results reveal a critical role for the XIAP-RIP2 interaction in NOD2 inflammatory signaling and provide a molecular basis for the design of innovative therapeutic strategies based on XIAP antagonists and RIP2 kinase inhibitors.


Subject(s)
Aminoquinolines/pharmacology , Inflammation/prevention & control , Nod2 Signaling Adaptor Protein/antagonists & inhibitors , Protein Interaction Domains and Motifs/drug effects , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Sulfones/pharmacology , X-Linked Inhibitor of Apoptosis Protein/metabolism , Animals , Cells, Cultured , Humans , Inflammation/metabolism , Inflammation/pathology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Signal Transduction , Ubiquitin/metabolism , Ubiquitination , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors
6.
Immunity ; 42(2): 321-331, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680273

ABSTRACT

T helper 1 (Th1) cell-associated immunity exacerbates ileitis induced by oral Toxoplasma gondii infection. We show here that attenuated ileitis observed in interleukin-22 (IL-22)-deficient mice was associated with reduced production of Th1-cell-promoting IL-18. IL-22 not only augmented the expression of Il18 mRNA and inactive precursor protein (proIL-18) in intestinal epithelial cells after T. gondii or Citrobacter rodentium infection, but also maintained the homeostatic amount of proIL-18 in the ileum. IL-22, however, did not induce the processing to active IL-18, suggesting a two-step regulation of IL-18 in these cells. Although IL-18 exerted pathogenic functions during ileitis triggered by T. gondii, it was required for host defense against C. rodentium. Conversely, IL-18 was required for the expression of IL-22 in innate lymphoid cells (ILCs) upon T. gondii infection. Our results define IL-18 as an IL-22 target gene in epithelial cells and describe a complex mutual regulation of both cytokines during intestinal infection.


Subject(s)
Enterobacteriaceae Infections/immunology , Interleukin-18/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Toxoplasmosis/immunology , Animals , Cells, Cultured , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/microbiology , Epithelial Cells/immunology , Ileitis/immunology , Ileitis/microbiology , Ileitis/parasitology , Ileum/immunology , Ileum/microbiology , Ileum/parasitology , Inflammation/immunology , Interferon-gamma/biosynthesis , Interleukin-18/biosynthesis , Interleukins/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Organ Culture Techniques , RNA, Messenger/biosynthesis , Th1 Cells/immunology , Toxoplasma/immunology , Toxoplasmosis/parasitology , Up-Regulation , Interleukin-22
7.
Nat Immunol ; 12(12): 1238-45, 2011 Oct 16.
Article in English | MEDLINE | ID: mdl-22001828

ABSTRACT

Interleukin 22 (IL-22), which is produced by cells of the T(H)17 subset of helper T cells and other leukocytes, not only enhances proinflammatory innate defense mechanisms in epithelial cells but also provides crucial protection to tissues from damage caused by inflammation and infection. In T(H)17 cells, transforming growth factor-ß (TGF-ß) regulates IL-22 and IL-17 differently. IL-6 alone induces T cells to produce only IL-22, whereas the combination of IL-6 and high concentrations of TGF-ß results in the production of IL-17 but not IL-22 by T cells. Here we identify the transcription factor c-Maf, which is induced by TGF-ß, as a downstream repressor of Il22. We found that c-Maf bound to the Il22 promoter and was both necessary and sufficient for the TGF-ß-dependent suppression of IL-22 production in T(H)17 cells.


Subject(s)
Interleukins/biosynthesis , Proto-Oncogene Proteins c-maf/metabolism , Th17 Cells/immunology , Transforming Growth Factor beta/pharmacology , Animals , Base Sequence , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Binding Sites/genetics , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Interleukins/genetics , Mice , Mice, Inbred BALB C , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Nucleotide Motifs , Promoter Regions, Genetic , Proto-Oncogene Proteins c-maf/genetics , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism , Th17 Cells/drug effects , Transcription, Genetic , Interleukin-22
8.
Nature ; 518(7539): 417-21, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25470037

ABSTRACT

T-helper type 17 (TH17) cells that produce the cytokines interleukin-17A (IL-17A) and IL-17F are implicated in the pathogenesis of several autoimmune diseases. The differentiation of TH17 cells is regulated by transcription factors such as RORγt, but post-translational mechanisms preventing the rampant production of pro-inflammatory IL-17A have received less attention. Here we show that the deubiquitylating enzyme DUBA is a negative regulator of IL-17A production in T cells. Mice with DUBA-deficient T cells developed exacerbated inflammation in the small intestine after challenge with anti-CD3 antibodies. DUBA interacted with the ubiquitin ligase UBR5, which suppressed DUBA abundance in naive T cells. DUBA accumulated in activated T cells and stabilized UBR5, which then ubiquitylated RORγt in response to TGF-ß signalling. Our data identify DUBA as a cell-intrinsic suppressor of IL-17 production.


Subject(s)
Interleukin-17/biosynthesis , Protein Biosynthesis , Th17 Cells/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Enzyme Stability , Female , Inflammation/genetics , Inflammation/pathology , Intestine, Small/metabolism , Intestine, Small/pathology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Signal Transduction , Substrate Specificity , Transforming Growth Factor beta/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/biosynthesis , Ubiquitin-Specific Proteases/deficiency , Ubiquitin-Specific Proteases/genetics , Ubiquitination
9.
Nature ; 514(7521): 237-41, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25119041

ABSTRACT

The connection between an altered gut microbiota and metabolic disorders such as obesity, diabetes, and cardiovascular disease is well established. Defects in preserving the integrity of the mucosal barriers can result in systemic endotoxaemia that contributes to chronic low-grade inflammation, which further promotes the development of metabolic syndrome. Interleukin (IL)-22 exerts essential roles in eliciting antimicrobial immunity and maintaining mucosal barrier integrity within the intestine. Here we investigate the connection between IL-22 and metabolic disorders. We find that the induction of IL-22 from innate lymphoid cells and CD4(+) T cells is impaired in obese mice under various immune challenges, especially in the colon during infection with Citrobacter rodentium. While innate lymphoid cell populations are largely intact in obese mice, the upregulation of IL-23, a cytokine upstream of IL-22, is compromised during the infection. Consequently, these mice are susceptible to C. rodentium infection, and both exogenous IL-22 and IL-23 are able to restore the mucosal host defence. Importantly, we further unveil unexpected functions of IL-22 in regulating metabolism. Mice deficient in IL-22 receptor and fed with high-fat diet are prone to developing metabolic disorders. Strikingly, administration of exogenous IL-22 in genetically obese leptin-receptor-deficient (db/db) mice and mice fed with high-fat diet reverses many of the metabolic symptoms, including hyperglycaemia and insulin resistance. IL-22 shows diverse metabolic benefits, as it improves insulin sensitivity, preserves gut mucosal barrier and endocrine functions, decreases endotoxaemia and chronic inflammation, and regulates lipid metabolism in liver and adipose tissues. In summary, we identify the IL-22 pathway as a novel target for therapeutic intervention in metabolic diseases.


Subject(s)
Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Immunity, Mucosal , Interleukins/immunology , Interleukins/metabolism , Metabolic Diseases/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chronic Disease , Citrobacter rodentium/drug effects , Citrobacter rodentium/immunology , Citrobacter rodentium/physiology , Colon/drug effects , Colon/immunology , Colon/microbiology , Diabetes Mellitus/pathology , Diet, High-Fat , Female , Hyperglycemia/diet therapy , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Immunity, Mucosal/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Insulin/metabolism , Insulin Resistance , Interleukin-23/immunology , Interleukin-23/metabolism , Interleukin-23/pharmacology , Interleukins/pharmacology , Interleukins/therapeutic use , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , Metabolic Diseases/diet therapy , Metabolic Diseases/drug therapy , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Receptors, Interleukin/deficiency , Receptors, Interleukin/metabolism , Receptors, Leptin/deficiency , Receptors, Leptin/metabolism , Interleukin-22
10.
Nature ; 509(7499): 235-9, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24739962

ABSTRACT

Reactive oxygen species (ROS) produced by phagocytes are essential for host defence against bacterial and fungal infections. Individuals with defective ROS production machinery develop chronic granulomatous disease. Conversely, excessive ROS can cause collateral tissue damage during inflammatory processes and therefore needs to be tightly regulated. Here we describe a protein, we termed negative regulator of ROS (NRROS), which limits ROS generation by phagocytes during inflammatory responses. NRROS expression in phagocytes can be repressed by inflammatory signals. NRROS-deficient phagocytes produce increased ROS upon inflammatory challenges, and mice lacking NRROS in their phagocytes show enhanced bactericidal activity against Escherichia coli and Listeria monocytogenes. Conversely, these mice develop severe experimental autoimmune encephalomyelitis owing to oxidative tissue damage in the central nervous system. Mechanistically, NRROS is localized to the endoplasmic reticulum, where it directly interacts with nascent NOX2 (also known as gp91(phox) and encoded by Cybb) monomer, one of the membrane-bound subunits of the NADPH oxidase complex, and facilitates the degradation of NOX2 through the endoplasmic-reticulum-associated degradation pathway. Thus, NRROS provides a hitherto undefined mechanism for regulating ROS production--one that enables phagocytes to produce higher amounts of ROS, if required to control invading pathogens, while minimizing unwanted collateral tissue damage.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Escherichia coli/immunology , Listeria monocytogenes/immunology , Proteins/metabolism , Reactive Oxygen Species/antagonists & inhibitors , Animals , Autoimmunity/genetics , Bone Marrow Cells/cytology , Central Nervous System/metabolism , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/metabolism , Female , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Latent TGF-beta Binding Proteins , Macrophages/cytology , Macrophages/enzymology , Macrophages/immunology , Macrophages/metabolism , Male , Membrane Proteins , Mice , NADPH Oxidases/metabolism , Oxidation-Reduction , Oxidative Stress , Phagocytes/cytology , Phagocytes/immunology , Phagocytes/metabolism , Proteins/genetics , Reactive Oxygen Species/metabolism
11.
Trends Immunol ; 36(8): 471-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26184648

ABSTRACT

The IL-1 and IL-10 family cytokines are important regulators of intestinal immunity. Whereas these cytokines have protective roles in response to mucosal damage or infection, they also contribute to pathology in certain settings. How these cytokines function to maintain intestinal homoeostasis, and under what circumstances they contribute to disease is poorly understood. Recent studies have revealed a multi-layered regulatory network wherein IL-1 and IL-10 family cytokines impact each other's production. The workings of this network vary in different intestinal regions, reflecting the influence of resident microbiota and the distribution of distinct immune cell populations in different regions of the intestine. We review these findings here, and discuss them in the context of the current understanding of the functions of these cytokine families in health and disease. We further highlight important areas of future investigation.


Subject(s)
Interleukin-10/immunology , Interleukin-1/immunology , Intestines/immunology , Animals , Humans
12.
Immunol Rev ; 252(1): 116-32, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23405899

ABSTRACT

Interleukin-22 (IL-22) has important functions in host defense at mucosal surfaces as well as in tissue repair. It is unique as a cytokine that is produced by immune cells, including T-helper (Th) cell subsets and innate lymphocytes, but acts only on non-hematopoietic stromal cells, in particular epithelial cells, keratinocytes, and hepatocytes. Although IL-22 is beneficial to the host in many infectious and inflammatory disorders, depending on the target tissue it can be pathogenic due to its inherent pro-inflammatory properties, which are further enhanced when IL-22 is released together with other pro-inflammatory cytokines, in particular IL-17. To avoid pathology, IL-22 and IL-17 production have to be controlled tightly and independently. While common factors such as signal transducer and activator of transcription 3 (STAT3) and retinoid orphan receptor γt (RORγt) drive the expression of both cytokines, other factors, such as c-Maf act specifically on IL-22 and enable the separate expression of either cytokine. Here, we discuss the production of IL-22 from various T-cell populations as well as protective versus pathogenic roles of IL-22. Finally, we focus on recent advances in our understanding of the molecular regulation of IL-22 in T cells.


Subject(s)
Interleukin-17/immunology , Interleukins/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Proto-Oncogene Proteins c-maf/immunology , STAT3 Transcription Factor/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Gene Expression Regulation , Humans , Immunity, Innate , Immunity, Mucosal , Interleukin-17/genetics , Interleukins/genetics , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Proto-Oncogene Proteins c-maf/genetics , STAT3 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes, Helper-Inducer/cytology , Interleukin-22
13.
Bioorg Med Chem Lett ; 26(18): 4455-4461, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27524313

ABSTRACT

Modification of the δ-sultam ring of RORc inverse agonist 2 led to the discovery of more polar oxa-sultam 65. The less lipophilic inverse agonist (65) displayed high potency in a biochemical assay, which translated into inhibition of IL-17 production in human peripheral blood mononuclear cells. The successful reduction of lipophilicity of this new analog gave rise to additional improvements in ROR selectivity and aqueous kinetic solubility, as well as reduction in plasma protein binding, while maintaining high cellular permeability.


Subject(s)
Lipids/chemistry , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Drug Discovery , Drug Inverse Agonism , Naphthalenesulfonates/chemistry
14.
Curr Top Microbiol Immunol ; 380: 213-36, 2014.
Article in English | MEDLINE | ID: mdl-25004820

ABSTRACT

Interleukin (IL)-22 is a member of the IL-10 family of cytokines, which, besides IL-10, contains seven additional cytokines. Although the founding member IL-10 is an important immunoregulatory cytokine that represses both innate and adaptive immunity, the other family members preferentially target epithelial cells and enhance innate host defense mechanisms against various pathogens such as bacteria, yeast, and viruses. Based on their functions, the IL-10 family can be further divided into three subgroups, IL-10 itself, the IL-20 subfamily, and the IFNλ subfamily. IL-22 is the best-studied member of the IL-20 subfamily, and exemplifies the diverse biological effects of this subfamily. IL-22 elicits various innate immune responses from epithelial cells and is essential for host defense against several invading pathogens, including Citrobacter rodentium and Klebsiella pneumonia. IL-22 also protects tissue integrity and maintains the mucosal homeostasis. On the other hand, IL-22 is a proinflammatory cytokine with the capacity to amplify inflammatory responses, which might result in tissue damage, e.g., the IL-22-dependent necrosis of the small intestine during Toxoplasma gondii infection.


Subject(s)
Infections/immunology , Interleukins/physiology , Mycoses/immunology , Animals , Bacterial Infections/immunology , Humans , Immunity, Innate , Interleukin-10/physiology , Toxoplasmosis/immunology , Interleukin-22
15.
Bioorg Med Chem Lett ; 25(15): 2907-12, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26048793

ABSTRACT

The nuclear receptor (NR) retinoic acid receptor-related orphan receptor gamma (RORγ, RORc, or NR1F3) is a promising target for the treatment of autoimmune diseases. RORc is a critical regulator in the production of the pro-inflammatory cytokine interleukin-17. We discovered a series of potent and selective imidazo[1,5-a]pyridine and -pyrimidine RORc inverse agonists. The most potent compounds displayed >300-fold selectivity for RORc over the other ROR family members, PPARγ, and NRs in our cellular selectivity panel. The favorable potency, selectivity, and physiochemical properties of GNE-0946 (9) and GNE-6468 (28), in addition to their potent suppression of IL-17 production in human primary cells, support their use as chemical biology tools to further explore the role of RORc in human biology.


Subject(s)
Imidazoles/chemistry , Imidazoles/pharmacology , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Cell Line , Cells, Cultured , Drug Discovery , HEK293 Cells , Humans , Imidazoles/metabolism , Imidazoles/pharmacokinetics , Interleukin-17/immunology , Liver/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Pyridines/metabolism , Pyridines/pharmacokinetics , Pyrimidines/metabolism , Pyrimidines/pharmacokinetics , Rats , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 24(16): 3891-7, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25017032

ABSTRACT

Using structure-based drug design principles, we identified opportunities to reduce the lipophilicity of our tertiary sulfonamide RORc inverse agonists. The new analogs possessed improved RORc cellular potencies with >77-fold selectivity for RORc over other nuclear receptors in our cell assay suite. The reduction in lipophilicity also led to an increased plasma-protein unbound fraction and improvements in cellular permeability and aqueous solubility.


Subject(s)
Blood Proteins/chemistry , Cell Membrane Permeability/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Sulfonamides/pharmacology , Animals , Binding Sites/drug effects , Blood Proteins/metabolism , Crystallography, X-Ray , Dogs , Dose-Response Relationship, Drug , Drug Design , Humans , Hydrophobic and Hydrophilic Interactions , Madin Darby Canine Kidney Cells , Models, Molecular , Molecular Structure , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Rats , Solubility , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
17.
Bioorg Med Chem Lett ; 24(24): 5769-5776, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25453817

ABSTRACT

The identification of a new series of RORc inverse agonists is described. Comprehensive structure-activity relationship studies of this reversed sulfonamide series identified potent RORc inverse agonists in biochemical and cellular assays which were also selective against a panel of nuclear receptors. Our work has contributed a compound that may serve as a useful in vitro tool to delineate the complex biological pathways involved in signalling through RORc. An X-ray co-crystal structure of an analogue with RORc has also provided useful insights into the binding interactions of the new series.


Subject(s)
Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Sulfonamides/chemistry , Binding Sites , Cell Survival/drug effects , Crystallography, X-Ray , Cytokines/biosynthesis , Drug Inverse Agonism , HEK293 Cells , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Molecular Dynamics Simulation , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/toxicity
18.
ACS Med Chem Lett ; 15(6): 864-872, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38894924

ABSTRACT

We were attracted to the therapeutic potential of inhibiting Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b), a RING E3 ligase that plays a critical role in regulating the activation of T cells. However, given that only protein-protein interactions were involved, it was unclear whether inhibition by a small molecule would be a viable approach. After screening an ∼6 billion member DNA-encoded library (DEL) using activated Cbl-b, we identified compound 1 as a hit for which the cis-isomer (2) was confirmed by biochemical and surface plasmon resonance (SPR) assays. Our hit optimization effort was greatly accelerated when we obtained a cocrystal structure of 2 with Cbl-b, which demonstrated induced binding at the substrate binding site, namely, the Src homology-2 (SH2) domain. This was quite noteworthy given that there are few reports of small molecule inhibitors that bind to SH2 domains and block protein-protein interactions. Structure- and property-guided optimization led to compound 27, which demonstrated measurable cell activity, albeit only at high concentrations.

19.
J Virol ; 86(3): 1802-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22114338

ABSTRACT

The downregulation of translation through eIF2α phosphorylation is a cellular response to diverse stresses, including viral infection, and is mediated by the GCN2 kinase, protein kinase R (PKR), protein kinase-like endoplasmic reticulum kinase (PERK), and heme-regulated inhibitor kinase (HRI). Although PKR plays a major role in defense against viruses, other eIF2α kinases also may respond to viral infection and contribute to the shutdown of protein synthesis. Here we describe the recessive, loss-of-function mutation atchoum (atc) in Eif2ak4, encoding GCN2, which increased susceptibility to infection by the double-stranded DNA viruses mouse cytomegalovirus (MCMV) and human adenovirus. This mutation was identified by screening macrophages isolated from mice carrying N-ethyl-N-nitrosourea (ENU)-induced mutations. Cells from Eif2ak4(atc/atc) mice failed to phosphorylate eIF2α in response to MCMV. Importantly, homozygous Eif2ak4(atc) mice showed a modest increase in susceptibility to MCMV infection, demonstrating that translational arrest dependent on GCN2 contributes to the antiviral response in vivo.


Subject(s)
DNA Virus Infections/genetics , Genetic Predisposition to Disease , Mutation , Protein Serine-Threonine Kinases/genetics , Animals , Base Sequence , DNA Primers , Mice , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction
20.
Blood ; 117(10): 2874-82, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21239699

ABSTRACT

Natural killer (NK) cells are innate immune cells that express members of the leukocyte ß2 integrin family in humans and mice. These CD11/CD18 heterodimers play critical roles in leukocyte trafficking, immune synapse formation, and costimulation. The cell-surface expression of one of these integrins, CD11b/CD18, is also recognized as a major marker of mouse NK-cell maturation, but its function on NK cells has been largely ignored. Using N-ethyl-N-nitrosourea (ENU) mutagenesis, we generated a mouse carrying an A → T transverse mutation in the Itgb2 gene, resulting in a mutation that prevented the cell-surface expression of CD18 and its associated CD11a, CD11b, and CD11c proteins. We show that ß2 integrin-deficient NK cells have a hyporesponsive phenotype in vitro, and present an alteration of their in vivo developmental program characterized by a selective accumulation of c-kit(+) cells. NK-cell missing-self recognition was partially altered in vivo, whereas the early immune response to mouse cytomegalovirus (MCMV) infection occurred normally in CD18-deficient mice. Therefore, ß2 integrins are required for optimal NK-cell maturation, but this deficiency is partial and can be bypassed during MCMV infection, highlighting the robustness of antiviral protective responses.


Subject(s)
CD18 Antigens/immunology , CD18 Antigens/metabolism , Cell Differentiation/immunology , Killer Cells, Natural/immunology , Animals , Cell Separation , Flow Cytometry , Herpesviridae Infections/immunology , Killer Cells, Natural/cytology , Mice , Muromegalovirus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL