Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 294(49): 18522-18531, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31578280

ABSTRACT

Cytokine macrophage migration inhibitory factor-2 (MIF-2 or D-dopachrome tautomerase) is a recently characterized second member of the MIF cytokine superfamily in mammalian genomes. MIF-2 shares pro-inflammatory and tumorigenic properties with the clinical target MIF (MIF-1), but the precise contribution of MIF-2 to immune physiology or pathology is unclear. Like MIF-1, MIF-2 has intrinsic keto-enol tautomerase activity and mediates biological functions by engaging the cognate, common MIF family receptor CD74. Evidence that the catalytic site of MIF family cytokines has a structural role in receptor binding has prompted exploration of tautomerase inhibitors as potential biological antagonists and therapeutic agents, although few catalytic inhibitors inhibit receptor activation. Here we describe the discovery and biochemical characterization of a selective small-molecule inhibitor of MIF-2. An in silico screen of 1.6 million compounds targeting the MIF-2 tautomerase site yielded several hits for potential catalytic inhibitors of MIF-2 and identified 4-(3-carboxyphenyl)-2,5-pyridinedicarboxylic acid (4-CPPC) as the most functionally potent compound. We found that 4-CPPC has an enzymatic IC50 of 27 µm and 17-fold selectivity for MIF-2 versus MIF-1. An in vitro binding assay for MIF-1/MIF-2 to the CD74 ectodomain (sCD74) indicated that 4-CPPC inhibits MIF-2-CD74 binding in a dose-dependent manner (0.01-10 µm) without influencing MIF-1-CD74 binding. Notably, 4-CPPC inhibited MIF-2-mediated activation of CD74 and reduced CD74-dependent signal transduction. These results open opportunities for development of more potent and pharmacologically auspicious MIF-2 inhibitors to investigate the distinct functions of this MIF family member in vivo.


Subject(s)
Intramolecular Oxidoreductases/metabolism , MSH Release-Inhibiting Hormone/metabolism , Humans , Inflammation/enzymology , Inflammation/metabolism , Intramolecular Oxidoreductases/chemistry , MSH Release-Inhibiting Hormone/chemistry , Neoplasms/enzymology , Neoplasms/metabolism , Protein Structure, Secondary , Signal Transduction
2.
Antioxid Redox Signal ; 31(14): 1053-1069, 2019 11 10.
Article in English | MEDLINE | ID: mdl-30767565

ABSTRACT

Significance: Pulmonary arterial hypertension (PAH) is a progressive disease arising from the narrowing of pulmonary arteries (PAs) resulting in high pulmonary arterial blood pressure and ultimately right ventricle (RV) failure. A defining characteristic of PAH is the excessive and unrelenting inward remodeling of PAs that includes increased proliferation, inflammation, and fibrosis. Critical Issues: There is no cure for PAH nor interventions that effectively arrest or reverse PA remodeling, and intensive research over the past several decades has sought to identify novel molecular mechanisms of therapeutic value. Recent Advances: Galectin-3 (Gal-3) is a carbohydrate-binding lectin remarkable for its chimeric structure, composed of an N-terminal oligomerization domain and a C-terminal carbohydrate-recognition domain. Gal-3 has been identified as a regulator of numerous changes in cell behavior that contributes to aberrant PA remodeling, including cell proliferation, inflammation, and fibrosis, but its role in PAH has remained poorly understood until recently. In contrast, pathological roles for Gal-3 have been proposed in cancer and inflammatory and fibroproliferative disorders, such as pulmonary vascular and cardiac fibrosis. Herein, we summarize the recent literature on the role of Gal-3 in the development of PAH. We provide experimental evidence supporting the ability of Gal-3 to influence reactive oxygen species production, NADPH oxidase enzyme expression, and redox signaling, which have been shown to contribute to both vascular remodeling and increased pulmonary arterial pressure. Future Directions: While several preclinical studies suggest that Gal-3 promotes hypertensive pulmonary vascular remodeling, the clinical significance of Gal-3 in human PAH remains to be established. Antioxid. Redox Signal. 00, 000-000.


Subject(s)
Fibrosis/metabolism , Galectin 3/metabolism , Inflammation/metabolism , Pulmonary Arterial Hypertension/metabolism , Reactive Oxygen Species/metabolism , Animals , Humans
3.
Am J Physiol Lung Cell Mol Physiol ; 316(5): L784-L797, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30724100

ABSTRACT

A defining characteristic of pulmonary hypertension (PH) is the extensive remodeling of pulmonary arteries (PAs), which results in progressive increases in vascular resistance and stiffness and eventual failure of the right ventricle. There is no cure for PH and identification of novel molecular mechanisms that underlie increased proliferation, reduced apoptosis, and excessive extracellular matrix production in pulmonary artery smooth muscle cells (PASMCs) is a vital objective. Galectin-3 (Gal-3) is a chimeric lectin and potent driver of many aspects of fibrosis, but its role in regulating PASMC behavior in PH remains poorly understood. Herein, we evaluated the importance of increased Gal-3 expression and signaling on PA vascular remodeling and cardiopulmonary function in experimental models of PH. Gal-3 expression was quantified by qRT-PCR, immunoblotting, and immunofluorescence imaging, and its functional role was assessed by specific Gal-3 inhibitors and CRISPR/Cas9-mediated knockout of Gal-3 in the rat. In rat models of PH, we observed increased Gal-3 expression in PASMCs, which stimulated migration and resistance to apoptosis, whereas silencing or genetic deletion reduced cellular migration and PA fibrosis and increased apoptosis. Gal-3 inhibitors attenuated and reversed PA remodeling and fibrosis, as well as hemodynamic indices in monocrotaline (MCT)-treated rats in vivo. These results were supported by genetic deletion of Gal-3 in both MCT and Sugen Hypoxia rat models. In conclusion, our results suggest that elevated Gal-3 levels contribute to inappropriate PA remodeling in PH by enhancing multiple profibrotic mechanisms. Therapeutic strategies targeting Gal-3 may be of benefit in the treatment of PH.


Subject(s)
Apoptosis , Cell Proliferation , Galectin 3/biosynthesis , Gene Expression Regulation , Hypertension, Pulmonary/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Fibrosis/metabolism , Animals , Blood Proteins , Disease Models, Animal , Galectin 3/genetics , Galectins , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Male , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Rats , Rats, Sprague-Dawley
5.
J Med Chem ; 61(3): 695-710, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29283260

ABSTRACT

This article highlights our work toward the identification of a potent, selective, and efficacious acidic mammalian chitinase (AMCase) inhibitor. Rational design, guided by X-ray analysis of several inhibitors bound to human chitotriosidase (hCHIT1), led to the identification of compound 7f as a highly potent AMCase inhibitor (IC50 values of 14 and 19 nM against human and mouse enzyme, respectively) and selective (>150× against mCHIT1) with very good PK properties. This compound dosed once daily at 30 mg/kg po showed significant anti-inflammatory efficacy in HDM-induced allergic airway inflammation in mice, reducing inflammatory cell influx in the BALF and total IgE concentration in plasma, which correlated with decrease of chitinolytic activity. Therapeutic efficacy of compound 7f in the clinically relevant aeroallergen-induced acute asthma model in mice provides a rationale for developing AMCase inhibitor for the treatment of asthma.


Subject(s)
Asthma/drug therapy , Asthma/enzymology , Chitinases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Molecular Targeted Therapy , Animals , CHO Cells , Chitinases/chemistry , Cricetulus , Disease Models, Animal , Enzyme Inhibitors/therapeutic use , Humans , Mice , Models, Molecular , Protein Conformation
6.
J Med Chem ; 56(6): 2568-80, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23472952

ABSTRACT

Recent efforts to identify treatments for myocardial ischemia reperfusion injury have resulted in the discovery of a novel series of highly potent α,α-disubstituted amino acid-based arginase inhibitors. The lead candidate, (R)-2-amino-6-borono-2-(2-(piperidin-1-yl)ethyl)hexanoic acid, compound 9, inhibits human arginases I and II with IC50s of 223 and 509 nM, respectively, and is active in a recombinant cellular assay overexpressing human arginase I (CHO cells). It is 28% orally bioavailable and significantly reduces the infarct size in a rat model of myocardial ischemia/reperfusion injury. Herein, we report the design, synthesis, and structure-activity relationships (SAR) for this novel series of inhibitors along with pharmacokinetic and in vivo efficacy data for compound 9 and X-ray crystallography data for selected lead compounds cocrystallized with arginases I and II.


Subject(s)
Amino Acids/chemistry , Amino Acids/pharmacology , Arginase/antagonists & inhibitors , Boron Compounds/chemistry , Boron Compounds/pharmacology , Caproates/chemistry , Caproates/pharmacology , Drug Discovery , Myocardial Reperfusion Injury/drug therapy , Amino Acids/pharmacokinetics , Amino Acids/therapeutic use , Animals , Arginase/chemistry , Boron Compounds/pharmacokinetics , Boron Compounds/therapeutic use , CHO Cells , Caproates/pharmacokinetics , Caproates/therapeutic use , Cricetinae , Cricetulus , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Male , Models, Molecular , Protein Conformation , Rats , Structure-Activity Relationship
7.
Biochem Biophys Res Commun ; 340(2): 482-90, 2006 Feb 10.
Article in English | MEDLINE | ID: mdl-16389067

ABSTRACT

Niacin is known to exert profound beneficial effects on cholesterol levels in humans, although its use is somewhat hampered by the gram quantities necessary to exert effects and the prevalence of compliance-limiting skin flushing side effects that occur. Recently, two G protein-coupled receptors (GPCRs) for niacin were identified and characterized as high (HM74A; GPR109A) and low (HM74; GPR109B) affinity receptors based on the binding affinities of niacin. These receptors also bind acifran (AY-25,712), which is known to modulate lipid levels like niacin, with similar affinities. Twelve analogs of acifran were chemically synthesized. One analogue demonstrated a dose-dependent decrease in serum triglycerides in rats within 3h of oral administration. Next, the acifran analogs were assessed for their activity towards the high and low affinity niacin receptors expressed in CHO-K1 cells. Constructs expressing HM74A or HM74 were stably transfected into CHO-K1 cells and shown to elicit phosphorylation of p42 and p44 mitogen-activated protein kinase (ERK1/ERK2) phosphorylation upon addition of niacin or acifran. The presence of functionally coupled GPCRs was further confirmed using Pertussis toxin, which completely inhibited the ability of either niacin or acifran to elicit phospho-ERK1/ERK2. The EC(50) of p-ERK1/ERK2 for niacin for the high and low affinity receptors was 47nM and indeterminate (i.e., >100microM), respectively, while the EC(50) for acifran was 160 and 316nM, respectively. Two chemical analogs of acifran demonstrated robust phosphorylation of ERK1/ERK2. Collectively, these data suggest that the synthesis of acifran analogs may be a suitable path for developing improved HM74A agonists.


Subject(s)
Furans/pharmacology , Lipid Metabolism/drug effects , Niacin/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Nicotinic/metabolism , Triglycerides/blood , Amino Acid Sequence , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Furans/administration & dosage , Furans/metabolism , Humans , Hypolipidemic Agents/pharmacology , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Sequence Data , Niacin/pharmacology , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, Nicotinic/biosynthesis , Receptors, Nicotinic/genetics
8.
J Immunol ; 172(3): 1391-6, 2004 Feb 01.
Article in English | MEDLINE | ID: mdl-14734714

ABSTRACT

IL-11 can reduce tissue injury in animal models of inflammation but the mechanism(s) is unknown. When C.B-17 SCID/beige mice bearing human skin grafts are injected i.p. with human PBMC allogeneic to the donor skin, infiltrating T cells destroy human microvessels by day 21. Intradermal injection of human IL-11 (500 ng/day) delays the time course of graft microvessel loss without reducing the extent of T cell infiltration. Protective actions of IL-11 are most pronounced on day 15. IL-11 has no effect on T cell activation marker, effector molecule, cytokine expression, or endothelial ICAM-1 expression. IL-11 up-regulates the expression of survivin, a cytoprotective protein, in graft keratinocytes and endothelial cells. Topical application of survivin antisense oligonucleotide down-regulates survivin expression in both cell types and largely abrogates the protective effect of IL-11. We conclude that in this human transplant model, IL-11 exerts a cytoprotective rather than anti-inflammatory or immunomodulatory effect mediated through induction of survivin.


Subject(s)
Cytoprotection/immunology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Interleukin-11/administration & dosage , Microtubule-Associated Proteins/biosynthesis , Skin Transplantation/pathology , Skin/blood supply , Skin/pathology , Adjuvants, Immunologic/administration & dosage , Administration, Topical , Adult , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Cytoprotection/genetics , Down-Regulation/genetics , Down-Regulation/immunology , Endothelium, Vascular/immunology , Humans , Inhibitor of Apoptosis Proteins , Injections, Intradermal , Interleukin-11/antagonists & inhibitors , Interleukin-11/therapeutic use , Leukocytes, Mononuclear/transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Microcirculation/immunology , Microcirculation/metabolism , Microcirculation/pathology , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Neoplasm Proteins , Ointments , Oligonucleotides, Antisense/administration & dosage , Skin/immunology , Skin/metabolism , Skin Transplantation/adverse effects , Skin Transplantation/immunology , Survivin , Transplantation, Homologous/adverse effects , Transplantation, Homologous/immunology , Transplantation, Homologous/pathology
9.
J Biol Chem ; 278(27): 25014-23, 2003 Jul 04.
Article in English | MEDLINE | ID: mdl-12724316

ABSTRACT

Oncostatin M (OnM) signals through cell surface receptors, which utilize the gp130 subunit. In cultured human umbilical vein endothelial cells (HUVEC), OnM transiently elevates mRNA encoding for suppressor of cytokine signaling-3 (SOCS-3). By 1 h of OnM treatment, HUVEC become refractory to the restimulation by OnM, measured as failure to reinduce SOCS-3 mRNA. OnM-induced desensitization also prevents responses to other gp130-signaling cytokines (e.g. leukemia inhibitory factor and interleukin 11). OnM treatment does not affect gp130 expression levels and desensitizes signaling mediated by a transduced chimeric receptor containing extracellular domains of platelet-derived growth factor receptor-beta (PDGFRbeta) and the cytoplasmic region of gp130. Interestingly, a chimeric PDGFRbeta-gp130 mutant receptor, in which intracellular Tyr residue 759 of gp130 is replaced by a Phe residue, mediates prolonged signaling and is not cross-desensitized by OnM. Phospho-Tyr759 is the binding site for both SOCS-3 and for Src homology domain 2-containing tyrosine phosphatase 2 (SHP-2). In human aortic smooth muscle cells, neither prevention of SOCS-3 protein induction, using STAT3 or SOCS-3 antisense, nor prevention of SHP-2 expression, also with antisense, ablates desensitization. These data suggest that desensitization of vascular cells to OnM is mediated in trans and involves Tyr residue 759 in gp130 but is not mediated by either SOCS-3 or SHP-2, the only two proteins currently known to bind to gp130 at this site.


Subject(s)
Antigens, CD/metabolism , Endothelium, Vascular/metabolism , Membrane Glycoproteins/metabolism , Peptides/metabolism , Protein Tyrosine Phosphatases/metabolism , Repressor Proteins , Transcription Factors , Animals , Antigens, CD/genetics , Cells, Cultured , Cytokine Receptor gp130 , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Mice , Oncostatin M , Protein Phosphatase 2 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Proteins/metabolism , SH2 Domain-Containing Protein Tyrosine Phosphatases , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Tyrosine , src Homology Domains
10.
Am J Respir Cell Mol Biol ; 29(4): 513-22, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12730073

ABSTRACT

Acute lung injury is a frequent and treatment-limiting consequence of therapy with 100% oxygen. Previous studies have determined that both interleukin (IL)-6 and IL-11 are protective in oxygen toxicity. This protection was associated with markedly diminished alveolar-capillary protein leak, endothelial and epithelial membrane injury, lipid peroxidation, and pulmonary neutrophil recruitment. Hyperoxia also caused cell death with DNA fragmentation in the lungs of transgene (-) animals, and both IL-6 and IL-11 markedly diminished this cell death response. However, the mechanism(s) by which these cytokines protect cells from death is unclear. In the present study, we characterized the effects of H2O2 on subconfluent human umbilical vein endothelial cell (HUVEC) and human pulmonary microvascular endothelial cell (HPMEC) cultures. We found that preincubation of HUVEC cultures with either IL-6 or IL-11 diminished H2O2 (1.0 mM)-induced cell death. Similar effects were noted with HPMEC showing that this effect is not HUVEC-specific. The protective effects of both IL-6 and IL-11 were not associated with any changes in antioxidants and were decreased by approximately 80% in the presence of U0126, a specific inhibitor of MEK-1-dependent pathways. The cytoprotective effects of IL-11 and IL-6 were also completely eliminated in STAT3 dominant-negative transduced HUVEC cultures. These studies demonstrate that IL-6 and IL-11 both confer cytoprotective effects that diminish oxidant-mediated endothelial cell injury. They also demonstrate that this protection is mediated, at least in part, by a STAT3 and MEK-1-dependent specific signal transduction pathway(s).


Subject(s)
Endothelium/drug effects , Hydrogen Peroxide/toxicity , Hyperoxia/chemically induced , Interleukin-11/pharmacology , Interleukin-6/pharmacology , Antioxidants/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endothelium/immunology , Endothelium/physiopathology , Humans , Hyperoxia/drug therapy , Hyperoxia/immunology , Interleukin-11/immunology , Interleukin-6/immunology , MAP Kinase Kinase 1 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Oxygen/toxicity , Pneumonia/chemically induced , Pneumonia/drug therapy , Pneumonia/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/immunology , STAT3 Transcription Factor , Signal Transduction/drug effects , Signal Transduction/immunology , Trans-Activators/genetics , Trans-Activators/metabolism
11.
J Biol Chem ; 277(10): 8012-21, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11777927

ABSTRACT

We compared human endothelial cell (EC) responses to interferon-gamma (IFN gamma) and oncostatin M (OnM), cytokines that utilize Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling. Both cytokines cause phosphorylation of Tyr residue 701 and Ser residue 727 of STAT1, as shown by immunoblotting. Both activate DNA binding of STAT1 homodimers, shown by electrophoretic mobility shift assay. However, only IFN gamma increases expression of three STAT1-dependent gene products examined, namely transporter associated with antigen processing-1 (TAP1), interferon regulatory factor-1 (IRF1), and class I major histocompatibility complex (MHC) protein, as demonstrated by immunoblotting. Only IFN gamma increases TAP1 transcription assessed by reporter gene assay. OnM pretreatment or co-treatment does not inhibit IFN gamma responses. Interestingly, IFN gamma activation of STAT1 is considerably more long-lived than that produced by OnM. To determine whether duration is functionally significant, we transduced EC with a chimeric receptor containing extracellular domains of platelet-derived growth factor receptor beta and intracellular regions of gp130, the signaling subunit of the OnM receptor, mutated to prevent binding of the tyrosine phosphatase SHP-2. Addition of platelet-derived growth factor to such transduced cells produces STAT1 activation that is comparable in magnitude and duration to that caused by IFN gamma, but still fails to induce TAP1, IRF1, or class I MHC molecules. OnM also activates STAT1 but not transcription of STAT1-dependent genes in HepG2 cells. Transient transfection of HepG2 cells with a STAT-defective mouse IFN gamma receptor failed to complement the OnM STAT signal. We conclude that STAT1 activation is necessary but not sufficient for induction of transcription of IFN gamma-responsive genes. However, signals provided by IFN gamma other than STAT1 activation cannot be provided in trans to complement the response to OnM.


Subject(s)
DNA-Binding Proteins/metabolism , Endothelium, Vascular/cytology , Peptides/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription, Genetic , Antineoplastic Agents/pharmacology , Cell Nucleus/metabolism , Cells, Cultured/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique, Indirect , Genes, Reporter , Green Fluorescent Proteins , Humans , Immunoblotting , Interferon-gamma/metabolism , Luminescent Proteins/metabolism , Oncostatin M , Peptides/pharmacology , Phosphorylation , Protein Binding , Recombinant Fusion Proteins/metabolism , Retroviridae/metabolism , STAT1 Transcription Factor , Transfection , Tyrosine/metabolism , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL