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.
Front Immunol ; 15: 1408772, 2024.
Article in English | MEDLINE | ID: mdl-38863703

ABSTRACT

Introduction: Macrophage dysfunction is a common feature of inflammatory disorders such as asthma, which is characterized by a strong circadian rhythm. Methods and results: We monitored the protein expression pattern of the molecular circadian clock in human peripheral blood monocytes from healthy, allergic, and asthmatic donors during a whole day. Monocytes cultured of these donors allowed us to examine circadian protein expression in human monocyte-derived macrophages, M1- and M2- polarized macrophages. In monocytes, particularly from allergic asthmatics, the oscillating expression of circadian proteins CLOCK, BMAL, REV ERBs, and RORs was significantly altered. Similar changes in BMAL1 were observed in polarized macrophages from allergic donors and in tissue-resident macrophages from activated precision cut lung slices. We confirmed clock modulating, anti-inflammatory, and lung-protective properties of the inverse ROR agonist SR1001 by reduced secretion of macrophage inflammatory protein and increase in phagocytosis. Using a house dust mite model, we verified the therapeutic effect of SR1001 in vivo. Discussion: Overall, our data suggest an interaction between the molecular circadian clock and monocytes/macrophages effector function in inflammatory lung diseases. The use of SR1001 leads to inflammatory resolution in vitro and in vivo and represents a promising clock-based therapeutic approach for chronic pulmonary diseases such as asthma.


Subject(s)
Asthma , Circadian Clocks , Macrophages , Monocytes , Humans , Monocytes/immunology , Monocytes/metabolism , Circadian Clocks/immunology , Animals , Macrophages/immunology , Macrophages/metabolism , Asthma/immunology , Asthma/metabolism , Male , Hypersensitivity/immunology , Hypersensitivity/metabolism , Inflammation/immunology , Female , Mice , Adult , Pyroglyphidae/immunology , Cells, Cultured , Circadian Rhythm/immunology
2.
Int J Mol Sci ; 22(21)2021 Nov 07.
Article in English | MEDLINE | ID: mdl-34769478

ABSTRACT

Placental hypervascularization has been reported in pregnancy-related pathologies such as gestational diabetes mellitus (GDM). Nevertheless, the underlying causes behind this abnormality are not well understood. In this study, we addressed the expression of SUCNR1 (cognate succinate receptor) in human placental endothelial cells and hypothesized that the succinate-SUCNR1 axis might play a role in the placental hypervascularization reported in GDM. We measured significantly higher succinate levels in placental tissue lysates from women with GDM relative to matched controls. In parallel, SUCNR1 protein expression was upregulated in GDM tissue lysates as well as in isolated diabetic fetoplacental arterial endothelial cells (FpECAds). A positive correlation of SUCNR1 and vascular endothelial growth factor (VEGF) protein levels in tissue lysates indicated a potential link between the succinate-SUCNR1 axis and placental angiogenesis. In our in vitro experiments, succinate prompted hallmarks of angiogenesis in human umbilical vein endothelial cells (HUVECs) such as proliferation, migration and spheroid sprouting. These results were further validated in fetoplacental arterial endothelial cells (FpECAs), where succinate induced endothelial tube formation. VEGF gene expression was increased in response to succinate in both HUVECs and FpECAs. Yet, knockdown of SUCNR1 in HUVECs led to suppression of VEGF gene expression and abrogated the migratory ability and wound healing in response to succinate. In conclusion, our data underline SUCNR1 as a promising metabolic target in human placenta and as a potential driver of enhanced placental angiogenesis in GDM.


Subject(s)
Neovascularization, Physiologic/genetics , Placenta/metabolism , Receptors, G-Protein-Coupled/metabolism , Adult , Case-Control Studies , Cells, Cultured , Diabetes, Gestational/genetics , Diabetes, Gestational/metabolism , Diabetes, Gestational/physiopathology , Endothelium, Vascular/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Placenta/blood supply , Pregnancy , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology
3.
Biochem Pharmacol ; 192: 114690, 2021 10.
Article in English | MEDLINE | ID: mdl-34274356

ABSTRACT

BACKGROUND: Eosinophilic asthma is increasingly recognized as one of the most severe and difficult-to-treat asthma subtypes. The JAK/STAT pathway is the principal signaling mechanism for a variety of cytokines and growth factors involved in asthma. However, the direct effect of JAK inhibitors on eosinophil effector function has not been addressed thus far. OBJECTIVE: Here we compared the effects of the JAK1/2 inhibitor baricitinib and the JAK3 inhibitor tofacitinib on eosinophil effector function in vitro and in vivo. METHODS: Differentiation of murine bone marrow-derived eosinophils. Migratory responsiveness, respiratory burst, phagocytosis and apoptosis of human peripheral blood eosinophils were assessed in vitro. In vivo effects were investigated in a mouse model of acute house dust mite-induced airway inflammation in BALB/c mice. RESULTS: Baricitinib more potently induced apoptosis and inhibited eosinophil chemotaxis and respiratory burst, while baricitinib and tofacitinib similarly affected eosinophil differentiation and phagocytosis. Of the JAK inhibitors, oral application of baricitinib more potently prevented lung eosinophilia in mice following allergen challenge. However, both JAK inhibitors neither affected airway resistance nor compliance. CONCLUSION: Our data suggest that the JAK1/2 inhibitor baricitinib is even more potent than the JAK3 inhibitor tofacitinib in suppressing eosinophil effector function. Thus, targeting the JAK1/2 pathway represents a promising therapeutic strategy for eosinophilic inflammation as observed in severe eosinophilic asthma.


Subject(s)
Azetidines/therapeutic use , Eosinophilia/drug therapy , Eosinophils/drug effects , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Janus Kinase Inhibitors/therapeutic use , Purines/therapeutic use , Pyrazoles/therapeutic use , Sulfonamides/therapeutic use , Adult , Animals , Azetidines/pharmacology , Cells, Cultured , Eosinophilia/chemically induced , Eosinophilia/immunology , Eosinophils/physiology , Female , Humans , Janus Kinase 1/immunology , Janus Kinase 2/immunology , Janus Kinase Inhibitors/pharmacology , Male , Mice , Mice, Inbred BALB C , Purines/pharmacology , Pyrazoles/pharmacology , Pyroglyphidae/immunology , Sulfonamides/pharmacology , Young Adult
4.
Allergy ; 75(2): 392-402, 2020 02.
Article in English | MEDLINE | ID: mdl-31408538

ABSTRACT

BACKGROUND: Recent studies pointed to a crucial role for apolipoproteins in the pathogenesis of inflammatory diseases. However, the role of apolipoprotein-IV (ApoA-IV) in allergic inflammation has not been addressed thoroughly thus far. OBJECTIVE: Here, we explored the anti-inflammatory effects and underlying signaling pathways of ApoA-IV on eosinophil effector function in vitro and in vivo. METHODS: Migratory responsiveness, Ca2+ -flux and apoptosis of human peripheral blood eosinophils were assessed in vitro. Allergen-driven airway inflammation was assessed in a mouse model of acute house dust mite-induced asthma. ApoA-IV serum levels were determined by ELISA. RESULTS: Recombinant ApoA-IV potently inhibited eosinophil responsiveness in vitro as measured by Ca2+ -flux, shape change, integrin (CD11b) expression, and chemotaxis. The underlying molecular mechanism involved the activation of Rev-ErbA-α and induced a PI3K/PDK1/PKA-dependent signaling cascade. Systemic application of ApoA-IV prevented airway hyperresponsiveness (AHR) and airway eosinophilia in mice following allergen challenge. ApoA-IV levels were decreased in serum from allergic patients compared to healthy controls. CONCLUSION: Our data suggest that ApoA-IV is an endogenous anti-inflammatory protein that potently suppresses effector cell functions in eosinophils. Thus, exogenously applied ApoA-IV may represent a novel pharmacological approach for the treatment of allergic inflammation and other eosinophil-driven disorders.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/blood , Apolipoproteins A/administration & dosage , Apolipoproteins A/blood , Asthma/blood , Asthma/drug therapy , Rhinitis/blood , Sinusitis/blood , Adolescent , Adult , Allergens/adverse effects , Animals , Anti-Inflammatory Agents/pharmacology , Apolipoproteins A/pharmacology , Apoptosis/drug effects , Asthma/etiology , Calcium/metabolism , Cells, Cultured , Chemotaxis/drug effects , Disease Models, Animal , Eosinophils/drug effects , Eosinophils/immunology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Pyroglyphidae/immunology , Young Adult
5.
Med Cannabis Cannabinoids ; 2(1): 14-21, 2019 Sep.
Article in English | MEDLINE | ID: mdl-34676329

ABSTRACT

The endocannabinoid system (ECS) is a multifunctional homeostatic system involved in many physiological and pathological conditions. The ligands of the ECS are the endo-cannabinoids, whose actions are mimicked by exogenous cannabinoids, such as phytocannabinoids and synthetic cannabinoids. Responses to the ligands of the ECS are mediated by numerous receptors like the classical cannabinoid receptors (CB1 and CB2) as well as ECS-related receptors, e.g., G protein-coupled receptors 18 and 55 (GPR18 and GPR55), transient receptor potential ion channels, and nuclear peroxisome proliferator-activated receptors. The ECS regulates almost all levels of female reproduction, starting with oocyte production through to parturition. Dysregulation of the ECS is associated with the development of gynecological disorders from fertility disorders to cancer. Cannabinoids that act at the ECS as specific agonists or antagonists may potentially influence dysregulation and, therefore, represent new therapeutic options for the therapy of gynecological disorders.

6.
J Allergy Clin Immunol ; 137(3): 833-43, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26792210

ABSTRACT

BACKGROUND: Prostaglandin (PG) D2 is an early-phase mediator in inflammation, but its action and the roles of the 2 D-type prostanoid receptors (DPs) DP1 and DP2 (also called chemoattractant receptor-homologous molecule expressed on T(H)2 cells) in regulating macrophages have not been elucidated to date. OBJECTIVE: We investigated the role of PGD2 receptors on primary human macrophages, as well as primary murine lung macrophages, and their ability to influence neutrophil action in vitro and in vivo. METHODS: In vitro studies, including migration, Ca(2+) flux, and cytokine secretion, were conducted with primary human monocyte-derived macrophages and neutrophils and freshly isolated murine alveolar and pulmonary interstitial macrophages. In vivo pulmonary inflammation was assessed in male BALB/c mice. RESULTS: Activation of DP1, DP2, or both receptors on human macrophages induced strong intracellular Ca(2+) flux, cytokine release, and migration of macrophages. In a murine model of LPS-induced pulmonary inflammation, activation of each PGD2 receptor resulted in aggravated airway neutrophilia, tissue myeloperoxidase activity, cytokine contents, and decreased lung compliance. Selective depletion of alveolar macrophages abolished the PGD2-enhanced inflammatory response. Activation of PGD2 receptors on human macrophages enhanced the migratory capacity and prolonged the survival of neutrophils in vitro. In human lung tissue specimens both DP1 and DP2 receptors were located on alveolar macrophages along with hematopoietic PGD synthase, the rate-limiting enzyme of PGD2 synthesis. CONCLUSION: For the first time, our results show that PGD2 markedly augments disease activity through its ability to enhance the proinflammatory actions of macrophages and subsequent neutrophil activation.


Subject(s)
Lung/immunology , Lung/metabolism , Macrophages/immunology , Macrophages/metabolism , Neutrophil Infiltration/immunology , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Animals , Calcium Signaling , Chemotaxis, Leukocyte/immunology , Cytokines/biosynthesis , Endotoxins/adverse effects , Endotoxins/immunology , Gene Expression , Humans , Inflammation Mediators/metabolism , Lung/pathology , Lung Injury/etiology , Lung Injury/metabolism , Lung Injury/pathology , Macrophages/drug effects , Mice , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Prostaglandin D2/pharmacology , Receptors, Immunologic/genetics , Receptors, Prostaglandin/genetics , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Tumor Necrosis Factor-alpha/biosynthesis
7.
Pharmacology ; 94(5-6): 280-6, 2014.
Article in English | MEDLINE | ID: mdl-25531811

ABSTRACT

Prostaglandin (PG) E2 has been implicated in the pathogenesis of aspirin-exacerbated respiratory disease (AERD). E-type prostanoid (EP) receptor 4 is known to confer inhibitory signals to eosinophils and monocytes, amongst others. In this study, we investigated whether the responsiveness of eosinophils and monocytes to PGE2 and EP4 receptor activation is altered in AERD patients. While the expression of the EP4 receptor in eosinophils was unaltered in AERD patients, inhibition of eosinophil chemotaxis by PGE2 or the EP4 agonist CAY10598 was less pronounced in AERD patients as compared to healthy control subjects. In monocytes, we found no changes in basal or lipopolysaccharide (LPS)-stimulated PGE2 synthesis, but the response to EP4 receptor activation with respect to inhibition of LPS-induced tumor necrosis factor-α release was reduced in AERD patients, especially in the presence of aspirin (acetylsalicylic acid). Our data point towards a decreased sensitivity of inhibitory EP4 receptor that may play a role in AERD.


Subject(s)
Aspirin/adverse effects , Asthma/chemically induced , Asthma/metabolism , Eosinophils/metabolism , Monocytes/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Adult , Aged , Aspirin/pharmacology , Calcium/metabolism , Cell Movement , Dinoprostone/metabolism , Dinoprostone/pharmacology , Eosinophils/physiology , Female , Humans , Lipopolysaccharides/pharmacology , Male , Middle Aged , Pyrrolidinones/pharmacology , Receptors, Prostaglandin E, EP4 Subtype/agonists , Tetrazoles/pharmacology , Tumor Necrosis Factor-alpha/metabolism
8.
Nat Chem Biol ; 8(7): 631-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22634634

ABSTRACT

Differential targeting of heterotrimeric G protein versus ß-arrestin signaling are emerging concepts in G protein-coupled receptor (GPCR) research and drug discovery, and biased engagement by GPCR ligands of either ß-arrestin or G protein pathways has been disclosed. Herein we report on a new mechanism of ligand bias to titrate the signaling specificity of a cell-surface GPCR. Using a combination of biomolecular and virtual screening, we identified the small-molecule modulator Gue1654, which inhibits Gßγ but not Gα signaling triggered upon activation of Gα(i)-ßγ by the chemoattractant receptor OXE-R in both recombinant and human primary cells. Gue1654 does not interfere nonspecifically with signaling directly at or downstream of Gßγ. This hitherto unappreciated mechanism of ligand bias at a GPCR highlights both a new paradigm for functional selectivity and a potentially new strategy to develop pathway-specific therapeutics.


Subject(s)
Benzeneacetamides/metabolism , Benzothiazoles/metabolism , Biopolymers/metabolism , GTP-Binding Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , Humans , Ligands
9.
Pharmacology ; 85(6): 372-82, 2010.
Article in English | MEDLINE | ID: mdl-20559016

ABSTRACT

Accumulation of type 2 T helper (Th2) lymphocytes and eosinophils is a hallmark of bronchial asthma and other allergic diseases, and it is believed that these cells play a crucial pathogenic role in allergic inflammation. Thus, Th2 cells and eosinophils are currently considered a major therapeutic target in allergic diseases and asthma. However, drugs that selectively target the accumulation and activation of Th2 cells and eosinophils in tissues are unavailable so far. Prostaglandin (PG)D(2) is a key mediator in various inflammatory diseases including allergy and asthma. It is generated by activated mast cells after allergen exposure and subsequently orchestrates the recruitment of inflammatory cells to the tissue. PGD(2) induces the chemotaxis of Th2 cells, basophils and eosinophils, stimulates cytokine release from these cells and prolongs their survival, and might hence indirectly promote IgE production. PGD(2) mediates its biologic functions via 2 distinct G protein-coupled receptors, D-type prostanoid receptor (DP), and the chemoattractant receptor-homologous molecule expressed on Th2 cells (CRTH2). DP and CRTH2 receptors are currently being considered as highly promising therapeutic targets for combating allergic diseases and asthma. Here, we revisit the roles of PGD(2) receptors in the regulation of eosinophil and Th2 cell function and the efforts towards developing candidate compounds for clinical evaluation.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Hypersensitivity, Immediate/drug therapy , Inflammation/drug therapy , Prostaglandin D2/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Prostaglandin/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Eosinophils/immunology , Eosinophils/metabolism , Humans , Hypersensitivity, Immediate/immunology , Hypersensitivity, Immediate/metabolism , Inflammation/immunology , Inflammation/metabolism , Mice , Receptors, Immunologic/metabolism , Receptors, Prostaglandin/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...