Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 15(2): e0228195, 2020.
Article in English | MEDLINE | ID: mdl-32053631

ABSTRACT

Tissue fibrosis is a pathological condition characterized by uncontrolled fibroblast activation that ultimately leads to organ failure. The TGFß1 pathway, one of the major players in establishment of the disease phenotype, is dependent on the transcriptional co-activators YAP/TAZ. We were interested whether fibroblasts can be sensitized to TGFß1 by activation of the GPCR/YAP/TAZ axis and whether this mechanism explains the profibrotic properties of diverse GPCR ligands. We found that LPA, S1P and thrombin cooperate in human dermal fibroblasts with TGFß1 to induce extracellular matrix synthesis, myofibroblast marker expression and cytokine secretion. Whole genome expression profiling identified a YAP/TAZ signature behind the synergistic profibrotic effects of LPA and TGFß1. LPA, S1P and thrombin stimulation led to activation of the Rho-YAP axis, an increase of nuclear YAP-Smad2 complexes and enhanced expression of profibrotic YAP/Smad2-target genes. More generally, dermal, cardiac and lung fibroblast responses to TGFß1 could be enhanced by increasing YAP nuclear levels (with GPCR ligands LPA, S1P, thrombin or Rho activator) and inhibited by decreasing nuclear YAP (with Rho inhibitor, forskolin, latrunculin B or 2-deoxy-glucose). Thus, we present here a conceptually interesting finding that fibroblast responses to TGFß1 can be predicted based on the nuclear levels of YAP and modulated by stimuli/treatments that change YAP nuclear levels. Our study contributes to better understanding of fibrosis as a complex interplay of signalling pathways and proposes YAP/TAZ as promising targets in the treatment of fibrosis.


Subject(s)
Cell Cycle Proteins/metabolism , Fibroblasts/pathology , Receptors, G-Protein-Coupled/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism , Cell Line , Enzyme Activation , Fibroblasts/metabolism , Fibrosis , Humans , Ligands , Lysophospholipids/metabolism , Signal Transduction , Smad2 Protein/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Thrombin/metabolism , rho-Associated Kinases/metabolism
2.
Am J Respir Cell Mol Biol ; 60(5): 578-591, 2019 05.
Article in English | MEDLINE | ID: mdl-30537446

ABSTRACT

Idiopathic pulmonary fibrosis is a life-threatening progressive disease characterized by loss of alveolar epithelial cells, inflammation, and aberrant fibroblast activation. The two currently approved therapies do not halt or reverse tissue remodeling, and therefore novel disease-modifying mechanisms are needed. Our results describe YAP/TAZ inhibition through prostacyclin (IP) receptor activation as a novel mechanism that suppresses profibrotic (myo)fibroblast activity. We investigated the antifibrotic properties of the selective IP receptor agonist ACT-333679 using primary human lung fibroblasts. ACT-333679 prevented transforming growth factor ß1-induced fibroblast-to-myofibroblast transition, proliferation, extracellular matrix synthesis, and IL-6 and PAI-1 secretion, and exerted relaxant effects in cell contraction assays. ACT-333679 treatment also reverted an established myofibroblast phenotype. Unbiased analysis of ACT-333679-induced whole-genome expression changes in transforming growth factor ß1-treated fibroblasts identified significant attenuation of genes regulated by YAP/TAZ, two transcriptional cofactors that are essential for fibrosis. We then demonstrated that ACT-333679, via elevation of cAMP, caused YAP/TAZ nuclear exclusion and subsequent suppression of YAP/TAZ-dependent profibrotic gene transcription. In summary, we offer a rationale for further exploring the potential of IP receptor agonists for the treatment of idiopathic pulmonary fibrosis.


Subject(s)
Acetates/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Fibroblasts/drug effects , Myofibroblasts/drug effects , Pyrazines/pharmacology , Receptors, Epoprostenol/genetics , Transcription Factors/genetics , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Case-Control Studies , Cell Differentiation , Cell Proliferation , Cyclic AMP/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/metabolism , Lung/pathology , Male , Myofibroblasts/metabolism , Myofibroblasts/pathology , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Epoprostenol/agonists , Receptors, Epoprostenol/metabolism , Signal Transduction , Transcription Factors/metabolism , Transcription, Genetic , Transforming Growth Factor beta1/pharmacology , YAP-Signaling Proteins
3.
J Pharmacol Exp Ther ; 362(1): 186-199, 2017 07.
Article in English | MEDLINE | ID: mdl-28476928

ABSTRACT

Prostacyclin (PGI2) receptor (IP receptor) agonists, which are indicated for the treatment of pulmonary arterial hypertension (PAH), increase cytosolic cAMP levels and thereby inhibit pulmonary vasoconstriction, pulmonary arterial smooth muscle cell (PASMC) proliferation, and extracellular matrix synthesis. Selexipag (Uptravi, 2-{4-[(5,6-diphenylpyrazin-2-yl)(isopropyl)amino]butoxy}-N-(methylsulfonyl)acetamide) is the first nonprostanoid IP receptor agonist, it is available orally and was recently approved for the treatment of PAH. In this study we show that the active metabolite of selexipag and the main contributor to clinical efficacy ACT-333679 (previously known as MRE-269) behaved as a full agonist in multiple PAH-relevant receptor-distal-or downstream-cellular assays with a maximal efficacy (Emax) comparable to that of the prototypic PGI2 analog iloprost. In PASMC, ACT-333679 potently induced cellular relaxation (EC50 4.3 nM) and inhibited cell proliferation (IC50 4.0 nM) as well as extracellular matrix synthesis (IC50 8.3 nM). In contrast, ACT-333679 displayed partial agonism in receptor-proximal-or upstream-cAMP accumulation assays (Emax 56%) when compared with iloprost and the PGI2 analogs beraprost and treprostinil (Emax ∼100%). Partial agonism of ACT-333679 also resulted in limited ß-arrestin recruitment (Emax 40%) and lack of sustained IP receptor internalization, whereas all tested PGI2 analogs behaved as full agonists in these desensitization-related assays. In line with these in vitro findings, selexipag, but not treprostinil, displayed sustained efficacy in rat models of pulmonary and systemic hypertension. Thus, the partial agonism of ACT-333679 allows for full efficacy in amplified receptor-distal PAH-relevant readouts while causing limited activity in desensitization-related receptor-proximal readouts.


Subject(s)
Acetamides/pharmacology , Acetates/pharmacology , Contractile Proteins/antagonists & inhibitors , Muscle Contraction/drug effects , Pyrazines/pharmacology , beta-Arrestins/metabolism , Animals , CHO Cells , Cell Proliferation/drug effects , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Epoprostenol/analogs & derivatives , Epoprostenol/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/physiopathology , Iloprost/pharmacology , Male , Muscle Relaxation/drug effects , Rats , Rats, Inbred SHR , Rats, Wistar , Receptors, Epoprostenol/agonists
4.
Mol Pharmacol ; 87(6): 916-27, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25762025

ABSTRACT

FTY720 phosphate (FTY720-P; 2-amino-2-[2-(4-octylphenyl)ethyl]-1,3-propanediol, monodihydrogen phosphate ester) is a nonselective sphingosine-1-phosphate (S1P) receptor agonist thought to be devoid of activity at the S1P2 receptor subtype. However, we have recently shown that FTY720-P displays significant S1P2 receptor agonist activity in recombinant cells and fibroblasts expressing endogenous S1P2 receptors. To elucidate the S1P2-dependent signaling pathways that were activated by FTY720-P, we employed second messenger assays and impedance-based assays in combination with pharmacological and small interfering RNA-based pathway inhibition in recombinant Chinese hamster ovary (CHO)-S1P2 cells as well as human lung myofibroblasts generated in vitro. In CHO-S1P2 cells, FTY720-P did not modulate cAMP or calcium levels. However, reporter-gene assays, impedance-based assays with a selective Rho-associated kinase (ROCK) inhibitor, Gα12/13 knockdown and activated Rho-pull-down assays demonstrated that FTY720-P potently activated Gα12/13/Rho/ROCK signaling. S1P similarly activated Gα12/13/Rho/ROCK signaling via S1P2 receptors, whereas the two selective S1P1 receptor agonists (Z,Z)-5-(3-chloro-4-[(2R)-2,3-dihydroxy-propoxy]-benzylidene)-2-propylimino-3-o-tolyl-thiazolidin-4-one (ponesimond) and 5-[4-phenyl-5-(trifluoromethyl)thiophen-2-yl]-3-[3-(trifluoromethyl)phenyl]1,2,4-oxadiazole (SEW2871) were inactive. In lung myofibroblasts, which mainly expressed the S1P2 receptor subtype, we showed that FTY720-P selectively activated the Gα12/13/Rho/ROCK pathway via the S1P2 receptor. Moreover, the activation of the Gα12/13/Rho/ROCK pathway in myofibroblasts by FTY720-P caused potent myofibroblast contraction similar to that induced by the natural ligand S1P. Thus, complementing second messenger assays with unbiased label-free assays or phenotypic assays in native expression systems can uncover activation of additional pathways, such as Gα12/13/Rho/ROCK signaling.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Myofibroblasts/drug effects , Organophosphates/pharmacology , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , rho-Associated Kinases/metabolism , Animals , CHO Cells , Calcium/metabolism , Cell Size/drug effects , Cricetinae , Cricetulus , Cyclic AMP/metabolism , Electric Impedance , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Genes, Reporter , Humans , Lung/cytology , Myofibroblasts/physiology , RNA, Small Interfering/genetics , Receptors, Lysosphingolipid/agonists , Signal Transduction , Sphingosine/pharmacology , rho-Associated Kinases/antagonists & inhibitors
5.
J Biol Chem ; 288(21): 14839-51, 2013 May 24.
Article in English | MEDLINE | ID: mdl-23589284

ABSTRACT

Synthetic sphingosine 1-phosphate receptor 1 modulators constitute a new class of drugs for the treatment of autoimmune diseases. Sphingosine 1-phosphate (S1P) signaling, however, is also involved in the development of fibrosis. Using normal human lung fibroblasts, we investigated the induction of fibrotic responses by the S1P receptor (S1PR) agonists S1P, FTY720-P, ponesimod, and SEW2871 and compared them with the responses induced by the known fibrotic mediator TGF-ß1. In contrast to TGF-ß1, S1PR agonists did not induce expression of the myofibroblast marker α-smooth muscle actin. However, TGF-ß1, S1P, and FTY720-P caused robust stimulation of extracellular matrix (ECM) synthesis and increased pro-fibrotic marker gene expression including connective tissue growth factor. Ponesimod showed limited and SEW2871 showed no pro-fibrotic potential in these readouts. Analysis of pro-fibrotic signaling pathways showed that in contrast to TGF-ß1, S1PR agonists did not activate Smad2/3 signaling but rather activated PI3K/Akt and ERK1/2 signaling to induce ECM synthesis. The strong induction of ECM synthesis by the nonselective agonists S1P and FTY720-P was due to the stimulation of S1P2 and S1P3 receptors, whereas the weaker induction of ECM synthesis at high concentrations of ponesimod was due to a low potency activation of S1P3 receptors. Finally, in normal human lung fibroblast-derived myofibroblasts that were generated by TGF-ß1 pretreatment, S1P and FTY720-P were effective stimulators of ECM synthesis, whereas ponesimod was inactive, because of the down-regulation of S1P3R expression in myofibroblasts. These data demonstrate that S1PR agonists are pro-fibrotic via S1P2R and S1P3R stimulation using Smad-independent pathways.


Subject(s)
Lung/metabolism , Lysophospholipids/metabolism , Myofibroblasts/metabolism , Organophosphates/pharmacology , Oxadiazoles/pharmacology , Pulmonary Fibrosis/metabolism , Receptors, Lysosphingolipid/agonists , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Thiophenes/pharmacology , Actins/genetics , Actins/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Down-Regulation/drug effects , Down-Regulation/genetics , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Humans , Lung/pathology , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myofibroblasts/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Fibrosis/genetics , Pulmonary Fibrosis/pathology , Receptors, Lysosphingolipid/biosynthesis , Receptors, Lysosphingolipid/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Sphingosine/antagonists & inhibitors , Sphingosine/genetics , Sphingosine/metabolism , Sphingosine/pharmacology , Thiazoles/pharmacology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
6.
J Med Chem ; 53(10): 4198-211, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20446681

ABSTRACT

Sphingosine-1-phosphate (S1P) is a widespread lysophospholipid which displays a wealth of biological effects. Extracellular S1P conveys its activity through five specific G-protein coupled receptors numbered S1P(1) through S1P(5). Agonists of the S1P(1) receptor block the egress of T-lymphocytes from thymus and lymphoid organs and hold promise for the oral treatment of autoimmune disorders. Here, we report on the discovery and detailed structure-activity relationships of a novel class of S1P(1) receptor agonists based on the 2-imino-thiazolidin-4-one scaffold. Compound 8bo (ACT-128800) emerged from this series and is a potent, selective, and orally active S1P(1) receptor agonist selected for clinical development. In the rat, maximal reduction of circulating lymphocytes was reached at a dose of 3 mg/kg. The duration of lymphocyte sequestration was dose dependent. At a dose of 100 mg/kg, the effect on lymphocyte counts was fully reversible within less than 36 h. Pharmacokinetic investigation of 8bo in beagle dogs suggests that the compound is suitable for once daily dosing in humans.


Subject(s)
Imines/chemical synthesis , Receptors, Lysosphingolipid/agonists , Thiazoles/chemical synthesis , Thiazolidines/chemical synthesis , Administration, Oral , Animals , Cell Line , Cricetinae , Cricetulus , Dogs , Hepatocytes/metabolism , High-Throughput Screening Assays , Humans , Imines/pharmacokinetics , Imines/pharmacology , Lymphocyte Count , Male , Microsomes, Liver/metabolism , Radioligand Assay , Rats , Rats, Wistar , Stereoisomerism , Structure-Activity Relationship , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Thiazolidines/pharmacokinetics , Thiazolidines/pharmacology
7.
J Cell Biochem ; 105(4): 1139-45, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18773427

ABSTRACT

Sphingosine-1-phosphate (S1P), acting through five closely related G-protein coupled receptors termed S1P1-5, has recently emerged as a possible regulator of smooth muscle cell (SMC) physiology with the potential to induce contraction, proliferation and stress fiber formation. In the present study, real-time quantitative PCR was used to determine the expression patterns of S1P receptor subtypes in human primary pulmonary artery smooth muscle cells (PASMC). We report here that subconfluent PASMC express predominantly S1P2 and S1P3 receptors and we show that S1P1 receptor mRNA levels are significantly up-regulated following basic fibroblast growth factor (bFGF) treatment. As a consequence, increased responsiveness, as measured by impedance and ERK1/2 phosphorylation, was observed upon stimulation with a specific S1P1 receptor agonist SEW2871. We therefore demonstrate, for the first time, that a growth factor that was previously shown to be involved in physiological and pathological changes of SMC function induced S1P1 receptor expression and we propose that S1P1 receptor up-regulation could contribute to vascular remodeling.


Subject(s)
Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/cytology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/physiology , Humans , RNA, Messenger/analysis , Receptors, Lysosphingolipid/analysis , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...