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.
J Cell Mol Med ; 15(11): 2430-42, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21199324

ABSTRACT

Airway smooth muscle cells exhibit phenotype plasticity that underpins their ability to contribute both to acute bronchospasm and to the features of airway remodelling in chronic asthma. A feature of mature, contractile smooth muscle cells is the presence of abundant caveolae, plasma membrane invaginations that develop from the association of lipid rafts with caveolin-1, but the functional role of caveolae and caveolin-1 in smooth muscle phenotype plasticity is unknown. Here, we report a key role for caveolin-1 in promoting phenotype maturation of differentiated airway smooth muscle induced by transforming growth factor (TGF)-ß(1). As assessed by Western analysis and laser scanning cytometry, caveolin-1 protein expression was selectively enriched in contractile phenotype airway myocytes. Treatment with TGF-ß(1) induced profound increases in the contractile phenotype markers sm-α-actin and calponin in cells that also accumulated abundant caveolin-1; however, siRNA or shRNAi inhibition of caveolin-1 expression largely prevented the induction of these contractile phenotype marker proteins by TGF-ß(1). The failure by TGF-ß(1) to adequately induce the expression of these smooth muscle specific proteins was accompanied by a strongly impaired induction of eukaryotic initiation factor-4E binding protein(4E-BP)1 phosphorylation with caveolin-1 knockdown, indicating that caveolin-1 expression promotes TGF-ß(1) signalling associated with myocyte maturation and hypertrophy. Furthermore, we observed increased expression of caveolin-1 within the airway smooth muscle bundle of guinea pigs repeatedly challenged with allergen, which was associated with increased contractile protein expression, thus providing in vivo evidence linking caveolin-1 expression with accumulation of contractile phenotype myocytes. Collectively, we identify a new function for caveolin-1 in controlling smooth muscle phenotype; this mechanism could contribute to allergic asthma.


Subject(s)
Caveolin 1/metabolism , Muscle Contraction , Myocytes, Smooth Muscle/metabolism , Respiratory System/metabolism , Actins/biosynthesis , Airway Remodeling , Animals , Asthma/physiopathology , Calcium-Binding Proteins , Caveolae/metabolism , Caveolae/physiology , Caveolin 1/genetics , Cells, Cultured , Dogs , Eukaryotic Initiation Factor-4E/metabolism , Guinea Pigs , Humans , Microfilament Proteins , Muscle Cells , Myocytes, Smooth Muscle/physiology , Phenotype , RNA Interference , RNA, Small Interfering , Signal Transduction , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Calponins
2.
Am J Respir Cell Mol Biol ; 44(3): 394-403, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20463291

ABSTRACT

Smooth muscle cells promote fibroproliferative airway remodeling in asthma, and transforming growth factor ß1 (TGFß1) is a key inductive signal. Statins are widely used to treat hyperlipidemia. Growing evidence indicates they also exert a positive impact on lung health, but the underlying mechanisms are unclear. We assessed the effects of 3-hydroxy-3-methlyglutaryl-coenzyme A (HMG-CoA) reductase inhibition with simvastatin on the fibrotic function of primary cultured human airway smooth muscle cells. Simvastatin blocked de novo cholesterol synthesis, but total myocyte cholesterol content was unaffected. Simvastatin also abrogated TGFß1-induced collagen I and fibronectin expression, and prevented collagen I secretion. The depletion of mevalonate cascade intermediates downstream from HMG-CoA underpinned the effects of simvastatin, because co-incubation with mevalonate, geranylgeranylpyrophosphate, or farnesylpyrophosphate prevented the inhibition of matrix protein expression. We also showed that human airway myocytes express both geranylgeranyl transferase 1 (GGT1) and farnesyltransferase (FT), and the inhibition of GGT1 (GGTI inhibitor-286, 10 µM), but not FT (FTI inhibitor-277, 10 µM), mirrored the suppressive effects of simvastatin on collagen I and fibronectin expression and collagen I secretion. Moreover, simvastatin and GGTI-286 both prevented TGFß1-induced membrane association of RhoA, a downstream target of GGT1. Our findings suggest that simvastatin and GGTI-286 inhibit synthesis and secretion of extracellular matrix proteins by human airway smooth muscle cells by suppressing GGT1-mediated posttranslational modification of signaling molecules such as RhoA. These findings reveal mechanisms related to evidence for the positive impact of statins on pulmonary health.


Subject(s)
Extracellular Matrix/metabolism , Gene Expression Regulation , Mevalonic Acid/metabolism , Trachea/metabolism , Transforming Growth Factor beta1/metabolism , Alkyl and Aryl Transferases/metabolism , Collagen/metabolism , Collagen Type I/metabolism , Farnesyltranstransferase/metabolism , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Models, Biological , Protein Processing, Post-Translational , Reverse Transcriptase Polymerase Chain Reaction , Simvastatin/pharmacology
3.
Am J Physiol Lung Cell Mol Physiol ; 294(6): L1110-8, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18390827

ABSTRACT

Beta-catenin plays a dual role in cellular signaling by stabilizing cadherin-mediated cell-cell contact and by regulating gene transcription associated with cell cycle progression. Nonetheless, its presence and function in airway smooth muscle have not been determined. We hypothesized a central role for beta-catenin in mitogenic signaling in airway smooth muscle in response to growth factor stimulation. Immunocytochemical and biochemical analysis revealed that human airway smooth muscle cells indeed express abundant beta-catenin, which was localized primarily to the plasma membrane in quiescent cells. Treatment of airway smooth muscle cells with PDGF or FBS induced sustained phosphorylation of glycogen synthase kinase-3 (GSK-3), a negative regulator in its unphosphorylated form that promotes beta-catenin degradation. GSK-3 phosphorylation was also increased in airway smooth muscle cells with a proliferative phenotype compared with quiescent airway smooth muscle cells with a mature phenotype. Parallel with the increase in GSK-3 phosphorylation, growth factor treatment induced an increased expression and nuclear presence of beta-catenin and activated promitogenic signaling in airway smooth muscle, including the phosphorylation of retinoblastoma protein, DNA synthesis ([(3)H]thymidine incorporation), and cell proliferation. Importantly, small interfering RNA knockdown of beta-catenin strongly reduced retinoblastoma protein phosphorylation, [(3)H]thymidine incorporation, and cell proliferation induced by PDGF and FBS. Collectively, these data reveal the existence of a GSK-3/beta-catenin signaling axis in airway smooth muscle that is regulated by growth factors and of central importance to mitogenic signaling.


Subject(s)
Glycogen Synthase Kinase 3/physiology , Muscle, Smooth/physiology , Signal Transduction/physiology , beta Catenin/physiology , Bronchi , Cell Line , Cell Proliferation/drug effects , Phosphorylation/drug effects , Platelet-Derived Growth Factor/pharmacology , RNA, Small Interfering/pharmacology , Retinoblastoma Protein/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 293(5): L1348-58, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17873004

ABSTRACT

Muscarinic receptors and platelet-derived growth factor (PDGF) receptors synergistically induce proliferation of airway smooth muscle (ASM), but the pathways that regulate these effects are not yet completely identified. We hypothesized that glycogen synthase kinase-3 (GSK-3), a kinase that represses several promitogenic signaling pathways in its unphosphorylated form, is cooperatively inhibited by PDGF and muscarinic receptors in immortalized human ASM cell lines. PDGF or methacholine alone induced rapid GSK-3 phosphorylation. This phosphorylation was sustained only for PDGF; however, methacholine potentiated PDGF-induced sustained GSK-3 phosphorylation. Synergistic effects of methacholine also were observed on PDGF-induced retinoblastoma protein (Rb) phosphorylation and cell proliferation. Suppression of GSK-3 inhibitory function using SB 216763 also augmented PDGF-induced Rb phosphorylation and cell cycle progression; this synergy was similar in magnitude to that seen for methacholine with PDGF. GSK-3 phosphorylation induced by methacholine required PKC, since it was abolished by GF 109203X and Gö 6976; however, inhibition of PKC had no effect on cell responses to PDGF. PKC inhibition also specifically abolished the synergistic effect of methacholine on PDGF-induced GSK-3 phosphorylation and cell proliferation. Collectively, these results show that GSK-3 plays a key repressive role in ASM cell proliferation. Moreover, muscarinic receptors mediate PKC-dependent GSK-3 inhibition, and this appears to be a primary mechanism underpinning augmentation of PDGF-induced cell growth.


Subject(s)
Cell Proliferation , Glycogen Synthase Kinase 3/metabolism , Myocytes, Smooth Muscle/physiology , Receptors, Muscarinic/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Respiratory System/metabolism , Blotting, Western , Cells, Cultured , Flow Cytometry , Gene Expression Regulation , Glycogen Synthase Kinase 3 beta , Humans , Immunoprecipitation , Methacholine Chloride/pharmacology , Muscarinic Agonists/pharmacology , Phosphorylation , Platelet-Derived Growth Factor/metabolism , Protein Kinase C/metabolism , Respiratory System/cytology , Retinoblastoma Protein , Signal Transduction
5.
Am J Physiol Lung Cell Mol Physiol ; 293(6): L1406-18, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17890325

ABSTRACT

Contractile responses of airway smooth muscle (ASM) determine airway resistance in health and disease. Caveolae microdomains in the plasma membrane are marked by caveolin proteins and are abundant in contractile smooth muscle in association with nanospaces involved in Ca(2+) homeostasis. Caveolin-1 can modulate localization and activity of signaling proteins, including trimeric G proteins, via a scaffolding domain. We investigated the role of caveolae in contraction and intracellular Ca(2+) ([Ca(2+)](i)) mobilization of ASM induced by the physiological muscarinic receptor agonist, acetylcholine (ACh). Human and canine ASM tissues and cells predominantly express caveolin-1. Muscarinic M(3) receptors (M(3)R) and Galpha(q/11) cofractionate with caveolin-1-rich membranes of ASM tissue. Caveolae disruption with beta-cyclodextrin in canine tracheal strips reduced sensitivity but not maximum isometric force induced by ACh. In fura-2-loaded canine and human ASM cells, exposure to methyl-beta-cyclodextrin (mbetaCD) reduced sensitivity but not maximum [Ca(2+)](i) induced by ACh. In contrast, both parameters were reduced for the partial muscarinic agonist, pilocarpine. Fluorescence microscopy revealed that mbetaCD disrupted the colocalization of caveolae-1 and M(3)R, but [N-methyl-(3)H]scopolamine receptor-binding assay revealed no effect on muscarinic receptor availability or affinity. To dissect the role of caveolin-1 in ACh-induced [Ca(2+)](i) flux, we disrupted its binding to signaling proteins using either a cell-permeable caveolin-1 scaffolding domain peptide mimetic or by small interfering RNA knockdown. Similar to the effects of mbetaCD, direct targeting of caveolin-1 reduced sensitivity to ACh, but maximum [Ca(2+)](i) mobilization was unaffected. These results indicate caveolae and caveolin-1 facilitate [Ca(2+)](i) mobilization leading to ASM contraction induced by submaximal concentrations of ACh.


Subject(s)
Calcium Signaling , Caveolae/metabolism , Intracellular Space/metabolism , Muscle Contraction/physiology , Muscle, Smooth/metabolism , Receptor, Muscarinic M3/metabolism , Respiratory System/metabolism , Acetylcholine/pharmacology , Animals , Calcium Signaling/drug effects , Caveolae/drug effects , Caveolin 1/chemistry , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Dogs , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Intracellular Space/drug effects , Muscle Cells/cytology , Muscle Cells/drug effects , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/ultrastructure , N-Methylscopolamine/metabolism , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Respiratory System/cytology , Respiratory System/drug effects , Respiratory System/ultrastructure , Trachea/cytology , Trachea/drug effects , Trachea/metabolism , Tritium/metabolism , beta-Cyclodextrins/pharmacology
6.
Am J Physiol Lung Cell Mol Physiol ; 292(5): L1163-72, 2007 May.
Article in English | MEDLINE | ID: mdl-17237147

ABSTRACT

Caveolae are abundant plasma membrane invaginations in airway smooth muscle that may function as preorganized signalosomes by sequestering and regulating proteins that control cell proliferation, including receptor tyrosine kinases (RTKs) and their signaling effectors. We previously demonstrated, however, that p42/p44 MAP kinase, a critical effector for cell proliferation, does not colocalize with RTKs in caveolae of quiescent airway myocytes. Therefore, we investigated the subcellular sites of growth factor-induced MAP kinase activation. In quiescent myocytes, though epidermal growth factor receptor (EGFR) was almost exclusively found in caveolae, p42/p44 MAP kinase, Grb2, and Raf-1 were absent from these membrane domains. EGF induced concomitant phosphorylation of caveolin-1 and p42/p44 MAP kinase; however, EGF did not promote the localization of p42/p44 MAP kinase, Grb2, or Raf-1 to caveolae. Interestingly, stimulation of muscarinic M(2) and M(3) receptors that were enriched in caveolae-deficient membranes also induced p42/p44 MAP kinase phosphorylation, but this occurred in the absence of caveolin-1 phosphorylation. This suggests that the localization of receptors to caveolae and interaction with caveolin-1 is not directly required for p42/p44 MAP kinase phosphorylation. Furthermore, we found that EGF exposure induced rapid translocation of EGFR from caveolae to caveolae-free membranes. EGFR trafficking coincided temporally with EGFR and p42/p44 MAP kinase phosphorylation. Collectively, this indicates that although caveolae sequester some receptors associated with p42/p44 MAP kinase activation, the site of its activation is associated with caveolae-free membrane domains. This reveals that directed trafficking of plasma membrane EGFR is an essential element of signal transduction leading to p42/p44 MAP kinase activation.


Subject(s)
Cell Membrane/physiology , ErbB Receptors/metabolism , Lung/enzymology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth/enzymology , Caveolae/physiology , Caveolin 1/analysis , Cell Division , Cell Line , Cell Membrane/ultrastructure , Clathrin/analysis , Humans , Receptors, Muscarinic/analysis
7.
Am J Physiol Lung Cell Mol Physiol ; 291(3): L523-34, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16617096

ABSTRACT

Chronic airways diseases, including asthma, are associated with an increased airway smooth muscle (ASM) mass, which may contribute to chronic airway hyperresponsiveness. Increased muscle mass is due, in part, to increased ASM proliferation, although the precise molecular mechanisms for this response are not completely clear. Caveolae, which are abundant in smooth muscle cells, are membrane microdomains where receptors and signaling effectors can be sequestered. We hypothesized that caveolae and caveolin-1 play an important regulatory role in ASM proliferation. Therefore, we investigated their role in p42/p44 MAPK signaling and proliferation using human ASM cell lines. Disruption of caveolae using methyl-beta-cyclodextrin and small interfering (si)RNA-knockdown of caveolin-1 caused spontaneous p42/p44 MAPK activation; additionally, caveolin-1 siRNA induced ASM proliferation in mitogen deficient conditions, suggesting a key role for caveolae and caveolin-1 in maintaining quiescence. Moreover, caveolin-1 accumulates twofold in myocytes induced to a contractile phenotype compared with proliferating ASM cells. Caveolin-1 siRNA failed to increase PDGF-induced p42/p44 MAPK activation and cell proliferation, however, indicating that PDGF stimulation actively reversed the antimitogenic control by caveolin-1. Notably, the PDGF induced loss of antimitogenic control by caveolin-1 coincided with a marked increase in caveolin-1 phosphorylation. Furthermore, the strong association of PDGF receptor-beta with caveolin-1 that exists in quiescent cells was rapidly and markedly reduced with agonist addition. This suggests a dynamic relationship in which mitogen stimulation actively reverses caveolin-1 suppression of p42/p44 MAPK signal transduction. As such, caveolae and caveolin-1 coordinate PDGF receptor signaling, leading to myocyte proliferation, and inhibit constitutive activity of p42/p44 MAPK to sustain cell quiescence.


Subject(s)
Caveolin 1/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth/metabolism , Caveolae/physiology , Caveolin 1/pharmacology , Cell Line , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enzyme Activation , Humans , Muscle Cells/physiology , Muscle, Smooth/cytology , Phosphorylation , Platelet-Derived Growth Factor , Receptor, Platelet-Derived Growth Factor beta/metabolism , Respiratory System , Signal Transduction , Telomerase/genetics , Telomerase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...