Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Circ Res ; 131(12): 1004-1017, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36321446

ABSTRACT

BACKGROUND: Genome-wide association studies have discovered a link between genetic variants on human chromosome 15q26.1 and increased coronary artery disease (CAD) susceptibility; however, the underlying pathobiological mechanism is unclear. This genetic locus contains the FES (FES proto-oncogene, tyrosine kinase) gene encoding a cytoplasmic protein-tyrosine kinase involved in the regulation of cell behavior. We investigated the effect of the 15q26.1 variants on FES expression and whether FES plays a role in atherosclerosis. METHODS AND RESULTS: Analyses of isogenic monocytic cell lines generated by CRISPR (clustered regularly interspaced short palindromic repeats)-mediated genome editing showed that monocytes with an engineered 15q26.1 CAD risk genotype had reduced FES expression. Small-interfering-RNA-mediated knockdown of FES promoted migration of monocytes and vascular smooth muscle cells. A phosphoproteomics analysis showed that FES knockdown altered phosphorylation of a number of proteins known to regulate cell migration. Single-cell RNA-sequencing revealed that in human atherosclerotic plaques, cells that expressed FES were predominately monocytes/macrophages, although several other cell types including smooth muscle cells also expressed FES. There was an association between the 15q26.1 CAD risk genotype and greater numbers of monocytes/macrophage in human atherosclerotic plaques. An animal model study demonstrated that Fes knockout increased atherosclerotic plaque size and within-plaque content of monocytes/macrophages and smooth muscle cells, in apolipoprotein E-deficient mice fed a high fat diet. CONCLUSIONS: We provide substantial evidence that the CAD risk variants at the 15q26.1 locus reduce FES expression in monocytes and that FES depletion results in larger atherosclerotic plaques with more monocytes/macrophages and smooth muscle cells. This study is the first demonstration that FES plays a protective role against atherosclerosis and suggests that enhancing FES activity could be a potentially novel therapeutic approach for CAD intervention.


Subject(s)
Atherosclerosis , Coronary Artery Disease , Plaque, Atherosclerotic , Proto-Oncogene Proteins c-fes , Animals , Humans , Mice , Arteries/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Genome-Wide Association Study , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Proto-Oncogene Proteins c-fes/genetics , Proto-Oncogene Proteins c-fes/metabolism
2.
Br J Pharmacol ; 179(21): 4958-4973, 2022 11.
Article in English | MEDLINE | ID: mdl-35802072

ABSTRACT

BACKGROUND AND PURPOSE: Vascular tone is regulated by the relative contractile state of vascular smooth muscle cells (VSMCs). Several integrins directly modulate VSMC contraction by regulating calcium influx through L-type voltage-gated Ca2+ channels (VGCCs). Genetic variants in ITGA9, which encodes the α9 subunit of integrin α9ß1, and SVEP1, a ligand for integrin α9ß1, associate with elevated blood pressure; however, neither SVEP1 nor integrin α9ß1 has reported roles in vasoregulation. We determined whether SVEP1 and integrin α9ß1 can regulate VSMC contraction. EXPERIMENTAL APPROACH: SVEP1 and integrin binding were confirmed by immunoprecipitation and cell binding assays. Human induced pluripotent stem cell-derived VSMCs were used in in vitro [Ca2+ ]i studies, and aortas from a Svep1+/- knockout mouse model were used in wire myography to measure vessel contraction. KEY RESULTS: We confirmed the ligation of SVEP1 to integrin α9ß1 and additionally found SVEP1 to directly bind to integrin α4ß1. Inhibition of SVEP1, integrin α4ß1 or α9ß1 significantly enhanced [Ca2+ ]i levels in isolated VSMCs to Gαq/11 -vasoconstrictors. This response was confirmed in whole vessels where a greater contraction to U46619 was seen in vessels from Svep1+/- mice compared to littermate controls or when integrin α4ß1 or α9ß1 was inhibited. Inhibition studies suggested that this effect was mediated via VGCCs, PKC and Rho A/Rho kinase dependent mechanisms. CONCLUSIONS AND IMPLICATIONS: Our studies reveal a novel role for SVEP1 and the integrins α4ß1 and α9ß1 in reducing VSMC contractility. This could provide an explanation for the genetic associations with blood pressure risk at the SVEP1 and ITGA9 loci.


Subject(s)
Induced Pluripotent Stem Cells , Integrin alpha4beta1 , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid , Animals , Calcium/metabolism , Cell Adhesion Molecules , Humans , Integrins/genetics , Integrins/metabolism , Ligands , Mice , Vasoconstriction , Vasoconstrictor Agents , rho-Associated Kinases
3.
Circulation ; 140(6): 500-513, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31163988

ABSTRACT

BACKGROUND: Genome-wide association studies have identified chromosome 14q32 as a locus for coronary artery disease. The disease-associated variants fall in a hitherto uncharacterized gene called HHIPL1 (hedgehog interacting protein-like 1), which encodes a sequence homolog of an antagonist of hedgehog signaling. The function of HHIPL1 and its role in atherosclerosis are unknown. METHODS: HHIPL1 cellular localization, interaction with sonic hedgehog (SHH), and influence on hedgehog signaling were tested. HHIPL1 expression was measured in coronary artery disease-relevant human cells, and protein localization was assessed in wild-type and Apoe-/- (apolipoprotein E deficient) mice. Human aortic smooth muscle cell phenotypes and hedgehog signaling were investigated after gene knockdown. Hhipl1-/- mice were generated and aortic smooth muscle cells collected for phenotypic analysis and assessment of hedgehog signaling activity. Hhipl1-/- mice were bred onto both the Apoe-/- and Ldlr-/- (low-density lipoprotein receptor deficient) knockout strains, and the extent of atherosclerosis was quantified after 12 weeks of high-fat diet. Cellular composition and collagen content of aortic plaques were assessed by immunohistochemistry. RESULTS: In vitro analyses revealed that HHIPL1 is a secreted protein that interacts with SHH and increases hedgehog signaling activity. HHIPL1 expression was detected in human smooth muscle cells and in smooth muscle within atherosclerotic plaques of Apoe-/- mice. The expression of Hhipl1 increased with disease progression in aortic roots of Apoe-/- mice. Proliferation and migration were reduced in Hhipl1 knockout mouse and HHIPL1 knockdown aortic smooth muscle cells, and hedgehog signaling was decreased in HHIPL1-deficient cells. Hhipl1 knockout caused a reduction of >50% in atherosclerosis burden on both Apoe-/- and Ldlr-/- knockout backgrounds, and lesions were characterized by reduced smooth muscle cell content. CONCLUSIONS: HHIPL1 is a secreted proatherogenic protein that enhances hedgehog signaling and regulates smooth muscle cell proliferation and migration. Inhibition of HHIPL1 protein function might offer a novel therapeutic strategy for coronary artery disease.


Subject(s)
Atherosclerosis/genetics , Chromosomes, Human, Pair 14/genetics , Coronary Disease/genetics , Hedgehog Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Animals , Atherosclerosis/pathology , Cell Division , Cell Movement , Cells, Cultured , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout, ApoE , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/pathology , Receptors, LDL/deficiency , Signal Transduction
4.
Eur Respir J ; 51(5)2018 05.
Article in English | MEDLINE | ID: mdl-29700102

ABSTRACT

Bronchial thermoplasty is a treatment for asthma. It is currently unclear whether its histopathological impact is sufficiently explained by the proportion of airway wall that is exposed to temperatures necessary to affect cell survival.Airway smooth muscle and bronchial epithelial cells were exposed to media (37-70°C) for 10 s to mimic thermoplasty. In silico we developed a mathematical model of airway heat distribution post-thermoplasty. In vivo we determined airway smooth muscle mass and epithelial integrity pre- and post-thermoplasty in 14 patients with severe asthma.In vitro airway smooth muscle and epithelial cell number decreased significantly following the addition of media heated to ≥65°C. In silico simulations showed a heterogeneous heat distribution that was amplified in larger airways, with <10% of the airway wall heated to >60°C in airways with an inner radius of ∼4 mm. In vivo at 6 weeks post-thermoplasty, there was an improvement in asthma control (measured via Asthma Control Questionnaire-6; mean difference 0.7, 95% CI 0.1-1.3; p=0.03), airway smooth muscle mass decreased (absolute median reduction 5%, interquartile range (IQR) 0-10; p=0.03) and epithelial integrity increased (14%, IQR 6-29; p=0.007). Neither of the latter two outcomes was related to improved asthma control.Integrated in vitro and in silico modelling suggest that the reduction in airway smooth muscle post-thermoplasty cannot be fully explained by acute heating, and nor did this reduction confer a greater improvement in asthma control.


Subject(s)
Asthma/therapy , Bronchial Thermoplasty/methods , Epithelial Cells/metabolism , Models, Biological , Muscle, Smooth/pathology , Adult , Aged , Airway Remodeling , Apoptosis , Bronchial Thermoplasty/adverse effects , Bronchoscopy , Computer Simulation , Female , Humans , Male , Middle Aged
5.
Arterioscler Thromb Vasc Biol ; 37(6): 1050-1057, 2017 06.
Article in English | MEDLINE | ID: mdl-28279971

ABSTRACT

OBJECTIVE: Genome-wide association studies have linked variants at chromosome 10q23 with increased coronary artery disease risk. The disease-associated variants fall in LIPA, which encodes lysosomal acid lipase (LAL), the enzyme responsible for lysosomal cholesteryl ester hydrolysis. Loss-of-function mutations in LIPA result in accelerated atherosclerosis. Surprisingly, the coronary artery disease variants are associated with increased LIPA expression in some cell types. In this study, we address this apparent contradiction. APPROACH AND RESULTS: We investigated a coding variant rs1051338, which is in high linkage disequilibrium (r2=0.89) with the genome-wide association study lead-associated variant rs2246833 and causes a nonsynonymous threonine to proline change within the signal peptide of LAL. Transfection of allele-specific expression constructs showed that the risk allele results in reduced lysosomal LAL protein (P=0.004) and activity (P=0.005). Investigation of LAL localization and turnover showed the risk LAL protein is degraded more quickly. This mechanism was confirmed in disease-relevant macrophages from individuals homozygous for either the nonrisk or risk allele. There was no difference in LAL protein or activity in whole macrophage extracts; however, we found reduced LAL protein (P=0.02) and activity (P=0.026) with the risk genotype in lysosomal extracts, suggesting that the risk genotype affects lysosomal LAL activity. Inhibition of the proteasome resulted in equal amounts of lysosomal LAL protein in risk and nonrisk macrophages. CONCLUSIONS: Our findings show that the coronary artery disease-associated coding variant rs1051338 causes reduced lysosomal LAL protein and activity because of increased LAL degradation, providing a plausible causal mechanism of increased coronary artery disease risk.


Subject(s)
Coronary Artery Disease/enzymology , Coronary Artery Disease/genetics , Lysosomes/enzymology , Macrophages/enzymology , Polymorphism, Single Nucleotide , Sterol Esterase/genetics , Sterol Esterase/metabolism , Adult , Animals , COS Cells , Chlorocebus aethiops , Down-Regulation , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Homozygote , Humans , Linkage Disequilibrium , Lysosomes/drug effects , Macrophages/drug effects , Male , Phenotype , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Protein Sorting Signals , Protein Transport , Proteolysis , Risk Factors , Transfection
6.
Proc Natl Acad Sci U S A ; 113(3): E291-9, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26733682

ABSTRACT

Protein transduction domains (PTDs) are powerful nongenetic tools that allow intracellular delivery of conjugated cargoes to modify cell behavior. Their use in biomedicine has been hampered by inefficient delivery to nuclear and cytoplasmic targets. Here we overcame this deficiency by developing a series of novel fusion proteins that couple a membrane-docking peptide to heparan sulfate glycosaminoglycans (GAGs) with a PTD. We showed that this GET (GAG-binding enhanced transduction) system could deliver enzymes (Cre, neomycin phosphotransferase), transcription factors (NANOG, MYOD), antibodies, native proteins (cytochrome C), magnetic nanoparticles (MNPs), and nucleic acids [plasmid (p)DNA, modified (mod)RNA, and small inhibitory RNA] at efficiencies of up to two orders of magnitude higher than previously reported in cell types considered hard to transduce, such as mouse embryonic stem cells (mESCs), human ESCs (hESCs), and induced pluripotent stem cells (hiPSCs). This technology represents an efficient strategy for controlling cell labeling and directing cell fate or behavior that has broad applicability for basic research, disease modeling, and clinical application.


Subject(s)
Cell-Penetrating Peptides/metabolism , Drug Delivery Systems , Glycosaminoglycans/metabolism , Amino Acid Motifs , Animals , Cell Differentiation/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Detergents/pharmacology , Endocytosis/drug effects , Genome , Homeodomain Proteins/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Integrases/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Muscle Development/drug effects , MyoD Protein/metabolism , NIH 3T3 Cells , Nanog Homeobox Protein , Nanoparticles , Nucleic Acids/metabolism , Protein Structure, Tertiary , Solubility , Trypsin/metabolism
7.
J Vis Exp ; (101): e52986, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26275100

ABSTRACT

Electrospinning is a highly adaptable method producing porous 3D fibrous scaffolds that can be exploited in in vitro cell culture. Alterations to intrinsic parameters within the process allow a high degree of control over scaffold characteristics including fiber diameter, alignment and porosity. By developing scaffolds with similar dimensions and topographies to organ- or tissue-specific extracellular matrices (ECM), micro-environments representative to those that cells are exposed to in situ can be created. The airway bronchiole wall, comprised of three main micro-environments, was selected as a model tissue. Using decellularized airway ECM as a guide, we electrospun the non-degradable polymer, polyethylene terephthalate (PET), by three different protocols to produce three individual electrospun scaffolds optimized for epithelial, fibroblast or smooth muscle cell-culture. Using a commercially available bioreactor system, we stably co-cultured the three cell-types to provide an in vitro model of the airway wall over an extended time period. This model highlights the potential for such methods being employed in in vitro diagnostic studies investigating important inter-cellular cross-talk mechanisms or assessing novel pharmaceutical targets, by providing a relevant platform to allow the culture of fully differentiated adult cells within 3D, tissue-specific environments.


Subject(s)
Bronchioles/cytology , Tissue Engineering/methods , Tissue Scaffolds , Adult , Cell Culture Techniques/methods , Cell Differentiation/physiology , Coculture Techniques/methods , Extracellular Matrix/physiology , Fibroblasts/cytology , Humans , Myocytes, Smooth Muscle/cytology , Polyethylene Terephthalates , Polymers , Porosity
8.
Biofabrication ; 6(3): 035003, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24722371

ABSTRACT

Electrospinning is a common technique used to fabricate fibrous scaffolds for tissue engineering applications. There is now growing interest in assessing the ability of collector plate design to influence the patterning of the fibres during the electrospinning process. In this study, we investigate a novel method to generate hybrid electrospun scaffolds consisting of both random fibres and a defined three-dimensional (3D) micro-topography at the surface, using patterned resin formers produced by rapid prototyping (RP). Poly(D,L-lactide-co-glycolide) was electrospun onto the engineered RP surfaces and the ability of these formers to influence microfibre patterning in the resulting scaffolds visualized by scanning electron microscopy. Electrospun scaffolds with patterns mirroring the microstructures of the formers were successfully fabricated. The effect of the resulting fibre patterns and 3D geometries on mammalian cell adhesion and proliferation was investigated by seeding enhanced green fluorescent protein labelled 3T3 fibroblasts onto the scaffolds. Following 24 h and four days of culture, the seeded scaffolds were visually assessed by confocal macro- and microscopy. The patterning of the fibres guided initial cell adhesion to the scaffold with subsequent proliferation over the geometry resulting in the cells being held in a 3D micro-topography. Such patterning could be designed to replicate a specific in vivo structure; we use the dermal papillae as an exemplar here. In conclusion, a novel, versatile and scalable method to produce hybrid electrospun scaffolds has been developed. The 3D directional cues of the patterned fibres have been shown to influence cell behaviour and could be used to culture cells within a similar 3D micro-topography as experienced in vivo.


Subject(s)
Electrochemical Techniques/methods , Lactic Acid/chemical synthesis , Polyglycolic Acid/chemical synthesis , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry , Animals , Cell Adhesion , Cell Proliferation , Cell Survival , Fibroblasts/cytology , Lactic Acid/chemistry , Mice , NIH 3T3 Cells , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer
9.
Am J Physiol Cell Physiol ; 302(5): C723-34, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22159081

ABSTRACT

Overstimulation of endothelin type A (ET(A)) and nucleotide (P2Y) Gα(q)-coupled receptors in vascular smooth muscle causes vasoconstriction, hypertension, and, eventually, hypertrophy and vascular occlusion. G protein-coupled receptor kinases (GRKs) and arrestin proteins are sequentially recruited by agonist-occupied Gα(q)-coupled receptors to terminate phospholipase C signaling, preventing prolonged/inappropriate contractile signaling. However, these proteins also play roles in the regulation of several mitogen-activated protein kinase (MAPK) signaling cascades known to be essential for vascular remodeling. Here we investigated whether different arrestin isoforms regulate endothelin and nucleotide receptor MAPK signaling in rat aortic smooth muscle cells (ASMCs). When intracellular Ca(2+) levels were assessed in isolated ASMCs loaded with Ca(2+)-sensitive dyes, P2Y(2) and ET(A) receptor desensitization was attenuated by selective small-interfering (si)RNA-mediated depletion of G protein-coupled receptor kinase 2 (GRK2). Using similar siRNA techniques, knockdown of arrestin2 prevented P2Y(2) receptor desensitization and enhanced and prolonged p38 and ERK MAPK signals, while arrestin3 depletion was ineffective. Conversely, arrestin3 knockdown prevented ET(A) receptor desensitization and attenuated ET1-stimulated p38 and ERK signals, while arrestin2 depletion had no effect. Using Transwell assays to assess agonist-stimulated ASMC migration, we found that UTP-stimulated migration was markedly attenuated following arrestin2 depletion, while ET1-stimulated migration was attenuated following knockdown of either arrestin. These data highlight a differential arrestin-dependent regulation of ET(A) and P2Y(2) receptor-stimulated MAPK signaling. GRK2 and arrestin expression are essential for agonist-stimulated ASMC migration, which, as a key process in vascular remodeling, highlights the potential roles of GRK2 and arrestin proteins in the progression of vascular disease.


Subject(s)
Arrestins/metabolism , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Receptor, Endothelin A/metabolism , Receptors, Purinergic P2Y2/metabolism , Animals , Arrestins/antagonists & inhibitors , Arrestins/genetics , Arteries/metabolism , Calcium/analysis , Cell Movement/physiology , Fura-2/analogs & derivatives , Fura-2/analysis , G-Protein-Coupled Receptor Kinase 2/metabolism , Gene Knockdown Techniques , Male , Mitogen-Activated Protein Kinases/metabolism , Myocytes, Smooth Muscle/chemistry , Myocytes, Smooth Muscle/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Signal Transduction , Type C Phospholipases/metabolism
10.
Cardiovasc Res ; 89(1): 193-203, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20705669

ABSTRACT

AIMS: prolonged P2Y-receptor signalling can cause vasoconstriction leading to hypertension, vascular smooth muscle hypertrophy, and hyperplasia. G protein-coupled receptor signalling is negatively regulated by G protein-coupled receptor kinases (GRKs) and arrestin proteins, preventing prolonged or inappropriate signalling. This study investigates whether GRKs and arrestins regulate uridine 5'-triphosphate (UTP)-stimulated contractile signalling in adult Wistar rat mesenteric arterial smooth muscle cells (MSMCs). METHODS AND RESULTS: mesenteric arteries contracted in response to UTP challenge: When an EC(50) UTP concentration (30 µM, 5 min) was added 5 min before (R(1)) and after (R(2)) the addition of a maximal UTP concentration (R(max): 100 µM, 5 min), R(2) responses were decreased relative to R(1), indicating desensitization. UTP-induced P2Y-receptor desensitization of phospholipase C signalling was studied in isolated MSMCs transfected with an inositol 1,4,5-trisphosphate biosensor and/or loaded with Ca(2+)-sensitive dyes. A similar protocol (R(1)/R(2) = 10 µM; R(max) = 100 µM, applied for 30 s) revealed markedly reduced R(2) when compared with R(1) responses. MSMCs were transfected with dominant-negative GRKs or siRNAs targeting specific GRK/arrestins to probe their respective roles in P2Y-receptor desensitization. GRK2 inhibition, but not GRK3, GRK5, or GRK6, attenuated P2Y-receptor desensitization. siRNA-mediated knockdown of arrestin2 attenuated UTP-stimulated P2Y-receptor desensitization, whereas arrestin3 depletion did not. Specific siRNA knockdown of the P2Y(2)-receptor almost completely abolished UTP-stimulated IP(3)/Ca(2+) signalling, strongly suggesting that our study is specifically characterizing this purinoceptor subtype. CONCLUSION: these new data highlight roles of GRK2 and arrestin2 as important regulators of UTP-stimulated P2Y(2)-receptor responsiveness in resistance arteries, emphasizing their potential importance in regulating vasoconstrictor signalling pathways implicated in vascular disease.


Subject(s)
Arrestins/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Purinergic P2Y/metabolism , Animals , Arrestins/antagonists & inhibitors , Arrestins/genetics , Base Sequence , Calcium Signaling , In Vitro Techniques , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth, Vascular/drug effects , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Receptors, Purinergic P2Y/classification , Receptors, Purinergic P2Y/genetics , Receptors, Purinergic P2Y2/genetics , Receptors, Purinergic P2Y2/metabolism , Signal Transduction , Uridine Triphosphate/metabolism , Uridine Triphosphate/pharmacology , beta-Arrestins
11.
Cardiovasc Res ; 85(3): 424-33, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19748906

ABSTRACT

AIMS: Prolonged endothelin (ET) receptor signalling causes vasoconstriction and can lead to hypertension, vascular smooth muscle hypertrophy, and hyperplasia. Usually, G protein-coupled receptor signalling is negatively regulated by G protein-coupled receptor kinases (GRKs), preventing prolonged or inappropriate signalling. This study investigated whether GRKs regulate ET receptor contractile signalling in adult Wistar rat mesenteric arterial smooth muscle cells (MSMCs). METHODS AND RESULTS: ET-1-stimulated phospholipase C (PLC) activity and changes in [Ca2+]i were assessed using confocal microscopy in rat MSMCs transfected with the pleckstrin-homology domain of PLCdelta1 (eGFP-PH) and loaded with Fura-Red. ET-1 applications (30 s) stimulated transient concentration-dependent eGFP-PH translocations from plasma membrane to cytoplasm and graded [Ca2+]i increases. ET-1-mediated PLC signalling was blocked by the type A endothelin receptor (ET(A)R) antagonist, BQ123. To characterize ET(A)R desensitization, cells were stimulated with a maximally effective concentration of ET-1 (50 nM, 30 s) followed by a variable washout period and a second identical application of ET-1. This brief exposure to ET-1 markedly decreased ET(A)R responsiveness to re-challenge, and reversal was incomplete even after increasing the time period between agonist challenges to 60 min. To assess GRK involvement in ET(A)R desensitization, MSMCs were co-transfected with eGFP-PH and catalytically inactive (D110A,K220R)GRK2, (D110A,K220R)GRK3, (K215R)GRK5, or (K215R)GRK6 constructs. (D110A,K220R)GRK2 expression significantly attenuated ET(A)R desensitization, whereas other constructs were ineffective. Small interfering RNA-targeted GRK2 depletion equally attenuated ET(A)R desensitization. Finally, immunocyotchemical data showed that ET(A)R activation recruited endogenous GRK2 from cytoplasm to membrane. CONCLUSION: These studies identify GRK2 as a key regulator of ET(A)R responsiveness in resistance arteries, highlighting the potential importance of this GRK isoenzyme in regulating vasoconstrictor signalling pathways implicated in vascular disease.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/physiology , Muscle, Smooth, Vascular/physiology , Receptor, Endothelin A/physiology , Signal Transduction/physiology , Animals , Calcium/metabolism , Cells, Cultured , Endothelin-1/pharmacology , G-Protein-Coupled Receptor Kinase 2/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Male , Muscle, Smooth, Vascular/enzymology , Protein Kinase C/physiology , Rats , Rats, Wistar , Vasoconstriction
12.
Mol Endocrinol ; 23(8): 1272-80, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19423652

ABSTRACT

Oxytocin plays an important role in the progression, timing, and modulation of uterine contraction during labor and is widely used as an uterotonic agent. We investigated the mechanisms regulating oxytocin receptor (OTR) signaling in human primary myometrial smooth muscle cells and the ULTR cell-line. Oxytocin produced concentration-dependent increases in both total [(3)H]inositol phosphate accumulation and intracellular Ca(2+) concentration ([Ca(2+)](i)); however, responses were greater and more reproducible in the ULTR cell line. Assessment of phospholipase C activity in single cells revealed that the OTR desensitizes rapidly (within 5 min) in the presence of oxytocin (100 nm). To characterize OTR desensitization further, cells were stimulated with a maximally effective concentration of oxytocin (100 nm, 30 sec) followed by a variable washout period and a second identical application of oxytocin. This brief exposure to oxytocin caused a marked decrease (>70%) in OTR responsiveness to rechallenge and was fully reversed by increasing the time period between agonist challenges. To assess involvement of G protein-coupled receptor kinases (GRKs) in OTR desensitization, cells were transfected with small interfering RNAs to cause specific > or =75% knockdown of GRKs 2, 3, 5, or 6. In both primary myometrial and ULTR cells, knockdown of GRK6 largely prevented oxytocin-induced OTR desensitization; in contrast, selective depletion of GRKs 2, 3, or 5 was without effect. These data indicate that GRK6 recruitment is a cardinal effector of OTR responsiveness and provide mechanistic insight into the likely in vivo regulation of OTR signaling in uterine smooth muscle.


Subject(s)
G-Protein-Coupled Receptor Kinases/metabolism , Gene Expression Regulation, Enzymologic , Muscle, Smooth/metabolism , Myometrium/metabolism , Receptors, Oxytocin/metabolism , Actins/metabolism , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Cell Line , Dose-Response Relationship, Drug , Female , Humans , Microfilament Proteins/metabolism , Models, Biological , Oxytocin/metabolism , Signal Transduction , Calponins
13.
FASEB J ; 20(12): 2153-5, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16935934

ABSTRACT

Viral and bacterial pathogens cause inflammation via Toll-like receptor (TLR) signaling. We have shown that effective responses to LPS may depend on cooperative interactions between TLR-expressing leukocytes and TLR-negative tissue cells. The aim of this work was to determine the roles of such networks in response to agonists of TLRs associated with antiviral and autoimmune responses. The TLR3 agonist poly(I:C) activated epithelial cells, primary endothelial cells, and two types of primary human smooth muscle cells (airway [ASMC] and vascular) directly, while the TLR7/8 agonist R848 required the presence of leukocytes to activate ASMC. In keeping with these data, ASMC expressed TLR3 but not TLR7 or TLR8. Activation of ASMC by poly(I:C) induced a specific cytokine repertoire characterized by induction of CXCL10 generation and the potential to recruit mast cells. We subsequently explored the ability of TLR agonists to cooperate in the induction of inflammation. Dual stimulation with LPS and poly(I:C) caused enhanced cytokine generation from epithelial and smooth muscle cells when in the presence of leukocytes. Thus, inflammatory responses to pathogens are regulated by networks in which patterns of TLR expression and colocalization of tissue cells and leukocytes are critical.


Subject(s)
Inflammation/etiology , Myocytes, Smooth Muscle/drug effects , Signal Transduction , Toll-Like Receptor 3/agonists , Toll-Like Receptors/physiology , Cells, Cultured , Coculture Techniques , Cytokines/biosynthesis , Endothelial Cells/cytology , Endothelial Cells/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Immunity , Inflammation/immunology , Leukocytes/cytology , Lipopolysaccharides/pharmacology , Myocytes, Smooth Muscle/cytology , Poly I-C/pharmacology , Toll-Like Receptor 3/physiology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 8/agonists , Toll-Like Receptors/agonists
14.
Am J Respir Crit Care Med ; 171(8): 814-22, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15657467

ABSTRACT

RATIONALE: Toll-like receptors 2 and 4 (TLR2, TLR4) enable cellular responses to bacterial lipoproteins, LPS, and endogenous mediators of cell damage. They have an established role in the activation of leukocytes, endothelial cells, and some smooth muscle cell types, but their roles in airway smooth muscle are uncertain. OBJECTIVES: To determine the roles of TLRs in activation of airway smooth muscle. METHODS: Airway smooth muscle cells were cultured with TLR agonists, in the presence or absence of mononuclear leukocytes. MEASUREMENTS AND MAIN RESULTS: We observed expression of TLR2 and TLR4 mRNAs, which could be upregulated by treatment with proinflammatory cytokines in primary human airway smooth muscle, but no important functional responses to agonists of these TLRs were seen. Coincubation of airway smooth muscle with peripheral blood mononuclear cells, at concentrations as low as 250 mononuclear cells/ml, resulted in a marked cooperative response to TLR stimuli, and synergistic production of cytokines, including chemokines (interleukin [IL-]-8) and IL-6. This cooperative response was greater when monocytes were enriched and was transferable using supernatants from LPS-stimulated peripheral blood mononuclear cells. Activation of cocultures required IL-1 generation from mononuclear cells, and was blocked by IL-1 receptor antagonist, though IL-1 generation alone was not sufficient to account for the magnitude of mononuclear cell-dependent coculture activation. CONCLUSIONS: These data indicate that potent amplification of inflammation induced by TLR agonists, such as LPS, may be achieved by cooperativity between airway smooth muscle and leukocytes involved in immune surveillance or inflammation.


Subject(s)
Lymphocyte Cooperation/drug effects , Muscle, Smooth/drug effects , Respiratory Muscles/drug effects , Toll-Like Receptor 2/agonists , Toll-Like Receptor 4/agonists , Cytokines/genetics , Cytokines/pharmacology , Escherichia coli , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Humans , In Vitro Techniques , Interleukin-1/genetics , Interleukin-1/pharmacology , Leukocyte Count , Lipopolysaccharides/immunology , Lymphocyte Cooperation/genetics , Muscle, Smooth/immunology , Neutrophils/drug effects , Neutrophils/immunology , RNA, Messenger/genetics , Respiratory Muscles/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Up-Regulation/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...