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










Publication year range
1.
Mol Pharmacol ; 88(1): 181-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25737495

ABSTRACT

G protein-coupled receptors (GPCRs), the largest family of signaling receptors in the human genome, are also the largest class of targets of approved drugs. Are the optimal GPCRs (in terms of efficacy and safety) currently targeted therapeutically? Especially given the large number (∼ 120) of orphan GPCRs (which lack known physiologic agonists), it is likely that previously unrecognized GPCRs, especially orphan receptors, regulate cell function and can be therapeutic targets. Knowledge is limited regarding the diversity and identity of GPCRs that are activated by endogenous ligands and that native cells express. Here, we review approaches to define GPCR expression in tissues and cells and results from studies using these approaches. We identify problems with the available data and suggest future ways to identify and validate the physiologic and therapeutic roles of previously unrecognized GPCRs. We propose that a particularly useful approach to identify functionally important GPCRs with therapeutic potential will be to focus on receptors that show selective increases in expression in diseased cells from patients and experimental animals.


Subject(s)
Gene Expression Profiling/methods , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , Gene Expression Regulation , Humans , Molecular Targeted Therapy , Receptors, G-Protein-Coupled/antagonists & inhibitors , Tissue Distribution
2.
J Biol Chem ; 289(42): 29148-57, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25183009

ABSTRACT

Epac, a guanine nucleotide exchange factor for the low molecular weight G protein Rap, is an effector of cAMP signaling and has been implicated to have roles in numerous diseases, including diabetes mellitus, heart failure, and cancer. We used a computational molecular modeling approach to predict potential binding sites for allosteric modulators of Epac and to identify molecules that might bind to these regions. This approach revealed that the conserved hinge region of the cyclic nucleotide-binding domain of Epac1 is a potentially druggable region of the protein. Using a bioluminescence resonance energy transfer-based assay (CAMYEL, cAMP sensor using YFP-Epac-Rluc), we assessed the predicted compounds for their ability to bind Epac and modulate its activity. We identified a thiobarbituric acid derivative, 5376753, that allosterically inhibits Epac activity and used Swiss 3T3 and HEK293 cells to test the ability of this compound to modulate the activity of Epac and PKA, as determined by Rap1 activity and vasodilator-stimulated phosphoprotein phosphorylation, respectively. Compound 5376753 selectively inhibited Epac in biochemical and cell migration studies. These results document the utility of a computational approach to identify a domain for allosteric regulation of Epac and a novel compound that prevents the activation of Epac1 by cAMP.


Subject(s)
Allosteric Site , Guanine Nucleotide Exchange Factors/metabolism , Animals , Cell Movement , Cell Survival , Computer Simulation , Cyclic AMP/metabolism , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Ligands , Mice , Molecular Dynamics Simulation , NIH 3T3 Cells , Protein Structure, Tertiary , Rats , Rats, Sprague-Dawley , Signal Transduction , Thiobarbituric Acid Reactive Substances
3.
Nat Commun ; 5: 4451, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-25043713

ABSTRACT

Progressive liver fibrosis is characterized by the deposition of collagen by activated hepatic stellate cells (HSCs). Activation of HSCs is a multiple receptor-driven process in which profibrotic signals are enhanced and antifibrotic pathways are suppressed. Here we report the discovery of a signalling platform comprising G protein subunit, Gαi and GIV, its guanine exchange factor (GEF), which serves as a central hub within the fibrogenic signalling network initiated by diverse classes of receptors. GIV is expressed in the liver after fibrogenic injury and is required for HSC activation. Once expressed, GIV enhances the profibrotic (PI3K-Akt-FoxO1 and TGFß-SMAD) and inhibits the antifibrotic (cAMP-PKA-pCREB) pathways to skew the signalling network in favour of fibrosis, all via activation of Gαi. We also provide evidence that GIV may serve as a biomarker for progression of fibrosis after liver injury and a therapeutic target for arresting and/or reversing HSC activation during liver fibrosis.


Subject(s)
Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Microfilament Proteins/metabolism , Vesicular Transport Proteins/metabolism , Animals , Cell Line , Collagen/biosynthesis , Guanine Nucleotide Exchange Factors/metabolism , Humans , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Up-Regulation , Vesicular Transport Proteins/genetics
4.
Am J Physiol Cell Physiol ; 306(9): C794-804, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24553187

ABSTRACT

Excess enzyme-mediated protein O-GlcNAcylation is known to occur with diabetes mellitus. A characteristic of diabetic cardiomyopathy is the development of myocardial fibrosis. The role that enhanced protein O-GlcNAcylation plays in modulating the phenotype of cardiac fibroblasts (CF) is unknown. To address this issue, rat CF were cultured in normal glucose (NG; 5 mM glucose) or high-glucose (HG; 25 mM) media for 48 h. Results demonstrate that CF cultured in HG have higher levels (~50%) of overall protein O-GlcNAcylation vs. NG cells. Key regulators of collagen synthesis such as transforming-growth factor-ß1 (TGF-ß1), SMADs 2/3, and SMAD 7 protein levels, including those of arginase I and II, were altered, leading to increases in collagen levels. The nuclear transcription factor Sp1 and arginase II evidence excess O-GlcNAcylation in HG cells. Expression in CF of an adenovirus coding for the enzyme N-acetylglucosaminidase, which removes O-GlcNAc moieties from proteins, decreased Sp1 and arginase II O-GlcNAcylation and restored HG-induced perturbations in CF back to NG levels. These findings may have important pathophysiological implications for the development of diabetes-induced cardiac fibrosis.


Subject(s)
Collagen/biosynthesis , Diabetic Cardiomyopathies/metabolism , Fibroblasts/metabolism , Glucose/metabolism , Myocardium/metabolism , Protein Processing, Post-Translational , Acetylglucosaminidase/genetics , Acetylglucosaminidase/metabolism , Animals , Arginase/metabolism , Cells, Cultured , Diabetic Cardiomyopathies/pathology , Fibroblasts/pathology , Glycosylation , Male , Myocardium/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction , Smad Proteins/metabolism , Sp1 Transcription Factor/metabolism , Time Factors , Transfection , Transforming Growth Factor beta1/metabolism , Up-Regulation
5.
Mol Pharmacol ; 84(6): 787-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24085841

ABSTRACT

Tissue fibrosis is characterized by excessive production, deposition, and contraction of the extracellular matrix (ECM). The second messenger cAMP has antifibrotic effects in fibroblasts from several tissues, including cardiac fibroblasts (CFs). Increased cellular cAMP levels can prevent the transformation of CFs into profibrogenic myofibroblasts, a critical step that precedes increased ECM deposition and tissue fibrosis. Here we tested two hypotheses: 1) myofibroblasts have a decreased ability to accumulate cAMP in response to G protein-coupled receptor (GPCR) agonists, and 2) increasing cAMP will not only prevent, but also reverse, the myofibroblast phenotype. We found that myofibroblasts produce less cAMP in response to GPCR agonists or forskolin and have decreased expression of several adenylyl cyclase (AC) isoforms and increased expression of multiple cyclic nucleotide phosphodiesterases (PDEs). Furthermore, we found that forskolin-promoted increases in cAMP or N(6)-phenyladenosine-cAMP, a protein kinase A-selective analog, reverse the myofibroblast phenotype, as assessed by the expression of collagen Iα1, α-smooth muscle actin, plasminogen activator inhibitor-1, and cellular contractile abilities, all hallmarks of a fibrogenic state. These results indicate that: 1) altered expression of AC and PDE isoforms yield a decrease in cAMP concentrations of cardiac myofibroblasts (relative to CFs) that likely contributes to their profibrotic state, and 2) approaches to increase cAMP concentrations not only prevent fibroblast-to-myofibroblast transformation but also can reverse the profibrotic myofibroblastic phenotype. We conclude that therapeutic strategies designed to enhance cellular cAMP concentrations in CFs may provide a means to reverse excessive scar formation following injury and to treat cardiac fibrosis.


Subject(s)
Cyclic AMP/biosynthesis , Myocardium/cytology , Myofibroblasts/cytology , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Actins/metabolism , Adenylyl Cyclases/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Collagen/biosynthesis , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis , Isoenzymes/metabolism , Male , Myocardium/metabolism , Myofibroblasts/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists
6.
Br J Pharmacol ; 166(2): 447-56, 2012 May.
Article in English | MEDLINE | ID: mdl-22233238

ABSTRACT

Fibrosis, the result of excess deposition of extracellular matrix (ECM), in particular collagen, leads to scarring and loss of function in tissues that include the heart, lung, kidney and liver. The second messenger cAMP can inhibit the formation and extent of ECM during this late phase of inflammation, but the mechanisms for these actions of cAMP and of agents that elevate tissue cAMP levels are not well understood. In this article, we review the fibrotic process and focus on two recently recognized aspects of actions of cAMP and its effector Epac (Exchange protein activated by cAMP): (a) blunting of epithelial-mesenchymal transformation (EMT) and (b) down-regulation of Epac expression by profibrotic agents (e.g. TGF-ß, angiotensin II), which may promote tissue fibrosis by decreasing Epac-mediated antifibrotic actions. Pharmacological approaches that raise cAMP or blunt the decrease in Epac expression by profibrotic agents may thus be strategies to block or perhaps reverse tissue fibrosis. LINKED ARTICLES This article is part of a themed section on Novel cAMP Signalling Paradigms. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.166.issue-2.


Subject(s)
Cyclic AMP/metabolism , Fibrosis/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Fibroblasts/metabolism , Humans
7.
J Mol Cell Cardiol ; 49(3): 362-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20471392

ABSTRACT

Cardiac fibroblasts (CFs) play a key role in response to injury and remodeling of the heart. Nucleotide (P2) receptors regulate the heart but limited information is available regarding such receptors in CFs. We thus sought to determine if extracellular nucleotides regulate fibrotic responses (e.g., proliferation, migration and expression of profibrotic markers) of CFs in primary culture. UTP increased rat CF migration 3-fold (p<0.001), proliferation by 30% (p<0.05) and mRNA expression of profibrotic markers: alpha smooth muscle actin (alpha-SMA), plasminogen activator inhibitor-1 (PAI-1), transforming growth factor beta, soluble ST2, interleukin-6 and monocyte chemoattractant protein-1 (MCP-1) by 3.0-, 15-, 2.0-, 7.6-, 11-, and 6.1-fold, respectively (p<0.05). PAI-1 protein expression induced by UTP was dependent on protein kinase C (PKC) and extracellular signal-regulated kinase (ERK), based on blockade by the PKC inhibitor Ro-31-8220 and the ERK inhibitor U0126, respectively. The rank order for enhanced expression of PAI-1 and alpha-SMA by nucleotides (UTPgammaS>>UDPbetaS>>ATPgammaS), the expression of P2Y2 receptors as the most abundantly expressed P2Y receptor in rat CFs and a blunted response to UTP in P2Y2(-/-) mice all implicate P2Y2 as the predominant P2Y receptor that mediates nucleotide-promoted profibrotic responses. Additional results indicate that P2Y2 receptor-promoted profibrotic responses in CFs are transient, perhaps as a consequence of receptor desensitization. We conclude that P2Y2 receptor activation is profibrotic in CFs; thus inhibition of P2Y2 receptors may provide a novel means to diminish fibrotic remodeling and turnover of extracellular matrix in the heart.


Subject(s)
Fibroblasts/drug effects , Heart/drug effects , Receptors, Purinergic P2Y2/metabolism , Uridine Triphosphate/pharmacology , Animals , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
8.
Proc Natl Acad Sci U S A ; 105(17): 6386-91, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18434542

ABSTRACT

Scar formation occurs during the late stages of the inflammatory response but, when excessive, produces fibrosis that can lead to functional and structural damage of tissues. Here, we show that the profibrogenic agonist, transforming growth factor beta1, transcriptionally decreases expression of Exchange protein activated by cAMP 1 (Epac1) in fibroblasts/fibroblast-like cells from multiple tissues (i.e., cardiac, lung, and skin fibroblasts and hepatic stellate cells). Overexpression of Epac1 inhibits transforming growth factor beta1-induced collagen synthesis, indicating that a decrease of Epac1 expression appears to be necessary for the fibrogenic phenotype, an idea supported by evidence that Epac1 expression in cardiac fibroblasts is inhibited after myocardial infarction. Epac and protein kinase A, a second mediator of cAMP action, have opposite effects on migration but both inhibit synthesis of collagen and DNA by fibroblasts. Epac is preferentially activated by low concentrations of cAMP and stimulates migration via the small G protein Rap1 but inhibits collagen synthesis in a Rap1-independent manner. The regulation of Epac expression and activation thus appear to be critical for the integration of pro- and anti-fibrotic signals and for the regulation of fibroblast function.


Subject(s)
Cyclic AMP/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Animals , Cell Movement/drug effects , Collagen Type I/biosynthesis , Collagen Type III/biosynthesis , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA/biosynthesis , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Male , Mice , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardium/enzymology , Myocardium/pathology , Phenotype , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , rap1 GTP-Binding Proteins/metabolism
9.
Am J Physiol Heart Circ Physiol ; 294(2): H736-49, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18024550

ABSTRACT

Sphingosine-1-phosphate (S1P) induces a transient bradycardia in mammalian hearts through activation of an inwardly rectifying K(+) current (I(K(ACh))) in the atrium that shortens action potential duration (APD) in the atrium. We have investigated probable mechanisms and receptor-subtype specificity for S1P-induced negative inotropy in isolated adult mouse ventricular myocytes. Activation of S1P receptors by S1P (100 nM) reduced cell shortening by approximately 25% (vs. untreated controls) in field-stimulated myocytes. S1P(1) was shown to be involved by using the S1P(1)-selective agonist SEW2871 on myocytes isolated from S1P(3)-null mice. However, in these myocytes, S1P(3) can modulate a somewhat similar negative inotropy, as judged by the effects of the S1P(1) antagonist VPC23019. Since S1P(1) activates G(i) exclusively, whereas S1P(3) activates both G(i) and G(q), these results strongly implicate the involvement of mainly G(i). Additional experiments using the I(K(ACh)) blocker tertiapin demonstrated that I(K(ACh)) can contribute to the negative inotropy following S1P activation of S1P(1) (perhaps through G(ibetagamma) subunits). Mathematical modeling of the effects of S1P on APD in the mouse ventricle suggests that shortening of APD (e.g., as induced by I(K(ACh))) can reduce L-type calcium current and thus can decrease the intracellular Ca(2+) concentration ([Ca(2+)](i)) transient. Both effects can contribute to the observed negative inotropic effects of S1P. In summary, these findings suggest that the negative inotropy observed in S1P-treated adult mouse ventricular myocytes may consist of two distinctive components: 1) one pathway that acts via G(i) to reduce L-type calcium channel current, blunt calcium-induced calcium release, and decrease [Ca(2+)](i); and 2) a second pathway that acts via G(i) to activate I(K(ACh)) and reduce APD. This decrease in APD is expected to decrease Ca(2+) influx and reduce [Ca(2+)](i) and myocyte contractility.


Subject(s)
Lysophospholipids/pharmacology , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Sphingosine/analogs & derivatives , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Depression, Chemical , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Heart Ventricles/cytology , Heart Ventricles/drug effects , In Vitro Techniques , Mice , Mice, Inbred C57BL , Models, Statistical , Potassium Channels, Inwardly Rectifying/drug effects , Receptors, G-Protein-Coupled/physiology , Receptors, Lysosphingolipid/drug effects , Receptors, Lysosphingolipid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Sphingosine/pharmacology
10.
Am J Physiol Heart Circ Physiol ; 293(1): H677-83, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17400715

ABSTRACT

Mechanical force can induce a number of fundamental short- and long-term responses in myocardium. These include alterations in ECM, activation of cell-signaling pathways, altered gene regulation, changes in cell proliferation and growth, and secretion of a number of peptides and growth factors. It is now known that a number of these autocrine/paracrine factors are secreted from both cardiomyocytes and ventricular cardiac fibroblasts (CFb) in response to stretch. One such substance is IGF-I. IGF-I is an important autocrine/paracrine factor that can regulate physiological or pathophysiological responses, such as hypertrophy. In this study, we addressed the possible effects of mechanical perturbation, biaxial strain, on IGF-I secretion from adult rat CFb. CFb were subjected to either static stretch (3-10%) or cyclic stretch (10%; 0.1-1 Hz) over a 24-h period. IGF-1 secretion from CFb in response to selected stretch paradigms was examined using ELISA to measure IGF-I concentrations in conditioned media. Static stretch did not result in any measurable modulation of IGF-I secretion from CFb. However, cyclic stretch significantly increased IGF-I secretion from CFb in a frequency- and time-dependent manner compared with nonstretched controls. This stretch-induced increase in secretion was relatively insensitive to changes in extracellular [Ca(2+)] or to block of L-type Ca(2+) channels. In contrast, thapsigargin, an inhibitor of sarco(endo)plasmic reticulum Ca(2+) ATPase, remarkably decreased stretch-induced IGF-I secretion from CFb. We further show that IGF-I can upregulate mRNA expression of atrial natriuretic peptide in myocytes. In summary, cyclic stretch can significantly increase IGF-I secretion from CFb, and this effect is dependent on a thapsigargin-sensitive pool of intracellular [Ca(2+)].


Subject(s)
Fibroblasts/physiology , Insulin-Like Growth Factor I/metabolism , Mechanotransduction, Cellular/physiology , Ventricular Function , Animals , Cells, Cultured , Elasticity , Male , Rats , Rats, Sprague-Dawley , Stress, Mechanical
11.
Am J Physiol Heart Circ Physiol ; 292(6): H2698-711, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17337593

ABSTRACT

The bioactive molecule sphingosine-1-phosphate (S1P) binds with high affinity to five recognized receptors (S1P(1-5)) to affect various tissues, including cellular responses of cardiac fibroblasts (CFbs) and myocytes. CFbs are essential components of myocardium, and detailed study of their cell signaling and physiology is required for a number of emerging disciplines. Meaningful studies on CFbs, however, necessitate methods for selective, reproducible cell isolations. Macrophages reside within normal cardiac tissues and often are isolated with CFbs. A protocol was therefore developed that significantly reduces macrophage levels and utilizes more CFb-specific markers (discoidin domain receptor-2) instead of, or in addition to, more commonly used cytoskeletal markers. Our results demonstrate that primary isolated, purified CFbs express predominantly S1P(1-3); however, the relative levels of these receptor subtypes are modulated with time and by culture conditions. In coculture experiments, macrophages altered CFb S1P receptor levels relative to controls. Further investigations using known macrophage-secreted factors showed that S1P and H(2)O(2) had minimal effects on CFb S1P(1-3) expression, whereas transforming growth factor-beta1, TNF-alpha, and PDGF-BB significantly altered all S1P receptor subtypes. Lowering FBS concentrations from 10% to 0.1% increased S1P(2), whereas supplementation with either PDGF-BB or Rho-associated protein kinase inhibitor Y-27632 significantly elevated S1P(3) levels. S1P(2) and S1P(3) receptor levels are known to regulate cell migration. Using cells isolated from either normal or S1P(3)-null mice, we demonstrate that S1P(3) is important and necessary for CFb migration. These results highlight the importance of demonstrating CFb culture purity in functional studies of S1P and also identify conditions that modulate S1P receptor expression in CFbs.


Subject(s)
Cell Culture Techniques , Cell Separation/methods , Fibroblasts/metabolism , Lysophospholipids/metabolism , Myocardium/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Amides/pharmacology , Animals , Becaplermin , Cell Communication , Cells, Cultured , Chemotaxis , Discoidin Domain Receptors , Fibroblasts/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/cytology , Phenotype , Platelet-Derived Growth Factor/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-sis , Pyridines/pharmacology , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Lysosphingolipid/deficiency , Receptors, Lysosphingolipid/genetics , Receptors, Mitogen/metabolism , Research Design , Serum/metabolism , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , Time Factors , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL