Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Respir Res ; 12: 113, 2011 Aug 24.
Article in English | MEDLINE | ID: mdl-21864337

ABSTRACT

BACKGROUND: Bronchial fibroblasts contribute to airway remodelling, including airway wall fibrosis. Transforming growth factor (TGF)-ß1 plays a major role in this process. We previously revealed the importance of the mevalonate cascade in the fibrotic response of human airway smooth muscle cells. We now investigate mevalonate cascade-associated signaling in TGFß1-induced fibronectin expression by bronchial fibroblasts from non-asthmatic and asthmatic subjects. METHODS: We used simvastatin (1-15 µM) to inhibit 3-hydroxy-3-methlyglutaryl-coenzyme A (HMG-CoA) reductase which converts HMG-CoA to mevalonate. Selective inhibitors of geranylgeranyl transferase-1 (GGT1; GGTI-286, 10 µM) and farnesyl transferase (FT; FTI-277, 10 µM) were used to determine whether GGT1 and FT contribute to TGFß1-induced fibronectin expression. In addition, we studied the effects of co-incubation with simvastatin and mevalonate (1 mM), geranylgeranylpyrophosphate (30 µM) or farnesylpyrophosphate (30 µM). RESULTS: Immunoblotting revealed concentration-dependent simvastatin inhibition of TGFß1 (2.5 ng/ml, 48 h)-induced fibronectin. This was prevented by exogenous mevalonate, or isoprenoids (geranylgeranylpyrophosphate or farnesylpyrophosphate). The effects of simvastatin were mimicked by GGTI-286, but not FTI-277, suggesting fundamental involvement of GGT1 in TGFß1-induced signaling. Asthmatic fibroblasts exhibited greater TGFß1-induced fibronectin expression compared to non-asthmatic cells; this enhanced response was effectively reduced by simvastatin. CONCLUSIONS: We conclude that TGFß1-induced fibronectin expression in airway fibroblasts relies on activity of GGT1 and availability of isoprenoids. Our results suggest that targeting regulators of isoprenoid-dependent signaling holds promise for treating airway wall fibrosis.


Subject(s)
Airway Remodeling/drug effects , Asthma/metabolism , Bronchi/drug effects , Fibroblasts/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Simvastatin/pharmacology , Transforming Growth Factor beta1/metabolism , Adult , Alkyl and Aryl Transferases/antagonists & inhibitors , Alkyl and Aryl Transferases/metabolism , Asthma/pathology , Bronchi/metabolism , Bronchi/pathology , Case-Control Studies , Cells, Cultured , Dose-Response Relationship, Drug , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibronectins/metabolism , Humans , Leucine/analogs & derivatives , Leucine/pharmacology , Methionine/analogs & derivatives , Methionine/pharmacology , Mevalonic Acid/metabolism , Polyisoprenyl Phosphates/metabolism , Sesquiterpenes/metabolism , Time Factors , Young Adult
2.
PLoS One ; 6(1): e16523, 2011 Jan 31.
Article in English | MEDLINE | ID: mdl-21304979

ABSTRACT

Statins inhibit the proximal steps of cholesterol biosynthesis, and are linked to health benefits in various conditions, including cancer and lung disease. We have previously investigated apoptotic pathways triggered by statins in airway mesenchymal cells, and identified reduced prenylation of small GTPases as a primary effector mechanism leading to p53-mediated cell death. Here, we extend our studies of statin-induced cell death by assessing endpoints of both apoptosis and autophagy, and investigating their interplay and coincident regulation. Using primary cultured human airway smooth muscle (HASM) and human airway fibroblasts (HAF), autophagy, and autophagosome formation and flux were assessed by transmission electron microscopy, cytochemistry (lysosome number and co-localization with LC3) and immunoblotting (LC3 lipidation and Atg12-5 complex formation). Chemical inhibition of autophagy increased simvastatin-induced caspase activation and cell death. Similarly, Atg5 silencing with shRNA, thus preventing Atg5-12 complex formation, increased pro-apoptotic effects of simvastatin. Simvastatin concomitantly increased p53-dependent expression of p53 up-regulated modulator of apoptosis (PUMA), NOXA, and damage-regulated autophagy modulator (DRAM). Notably both mevalonate cascade inhibition-induced autophagy and apoptosis were p53 dependent: simvastatin increased nuclear p53 accumulation, and both cyclic pifithrin-α and p53 shRNAi partially inhibited NOXA, PUMA expression and caspase-3/7 cleavage (apoptosis) and DRAM expression, Atg5-12 complex formation, LC3 lipidation, and autophagosome formation (autophagy). Furthermore, the autophagy response is induced rapidly, significantly delaying apoptosis, suggesting the existence of a temporally coordinated p53 regulation network. These findings are relevant for the development of statin-based therapeutic approaches in obstructive airway disease.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Mesoderm/cytology , Mevalonic Acid/pharmacology , Respiratory System/cytology , Tumor Suppressor Protein p53/physiology , Cells, Cultured , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mesoderm/drug effects , Respiratory System/drug effects , Simvastatin/pharmacology
3.
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
4.
J Cell Sci ; 123(Pt 18): 3061-70, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20736308

ABSTRACT

The dystrophin-glycoprotein complex (DGC) links the extracellular matrix and actin cytoskeleton. Caveolae form membrane arrays on smooth muscle cells; we investigated the mechanism for this organization. Caveolin-1 and beta-dystroglycan, the core transmembrane DGC subunit, colocalize in airway smooth muscle. Immunoprecipitation revealed the association of caveolin-1 with beta-dystroglycan. Disruption of actin filaments disordered caveolae arrays, reduced association of beta-dystroglycan and caveolin-1 to lipid rafts, and suppressed the sensitivity and responsiveness of methacholine-induced intracellular Ca2+ release. We generated novel human airway smooth muscle cell lines expressing shRNA to stably silence beta-dystroglycan expression. In these myocytes, caveolae arrays were disorganized, caveolae structural proteins caveolin-1 and PTRF/cavin were displaced, the signaling proteins PLCbeta1 and G(alphaq), which are required for receptor-mediated Ca2+ release, were absent from caveolae, and the sensitivity and responsiveness of methacholine-induced intracellular Ca2+ release, was diminished. These data reveal an interaction between caveolin-1 and beta-dystroglycan and demonstrate that this association, in concert with anchorage to the actin cytoskeleton, underpins the spatial organization and functional role of caveolae in receptor-mediated Ca2+ release, which is an essential initiator step in smooth muscle contraction.


Subject(s)
Calcium/metabolism , Caveolae/metabolism , Caveolin 1/metabolism , Dystroglycans/metabolism , Muscle, Smooth/metabolism , Animals , Caveolin 1/genetics , Cells, Cultured , Cytoskeleton/genetics , Cytoskeleton/metabolism , Dogs , Dystroglycans/genetics , Humans , Muscle Cells/metabolism , Protein Binding
5.
Biochim Biophys Acta ; 1803(4): 452-67, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20045437

ABSTRACT

Statins inhibit 3-hydroxy-3-methyl-glutarylcoenzyme CoA (HMG-CoA) reductase, the proximal enzyme for cholesterol biosynthesis. They exhibit pleiotropic effects and are linked to health benefits for diseases including cancer and lung disease. Understanding their mechanism of action could point to new therapies, thus we investigated the response of primary cultured human airway mesenchymal cells, which play an effector role in asthma and chronic obstructive lung disease (COPD), to simvastatin exposure. Simvastatin induced apoptosis involving caspase-9, -3 and -7, but not caspase-8 in airway smooth muscle cells and fibroblasts. HMG-CoA inhibition did not alter cellular cholesterol content but did abrogate de novo cholesterol synthesis. Pro-apoptotic effects were prevented by exogenous mevalonate, geranylgeranyl pyrophosphate and farnesyl pyrophosphate, downstream products of HMG-CoA. Simvastatin increased expression of Bax, oligomerization of Bax and Bak, and expression of BH3-only p53-dependent genes, PUMA and NOXA. Inhibition of p53 and silencing of p53 unregulated modulator of apoptosis (PUMA) expression partly counteracted simvastatin-induced cell death, suggesting a role for p53-independent mechanisms. Simvastatin did not induce mitochondrial release of cytochrome c, but did promote release of inhibitor of apoptosis (IAP) proteins, Smac and Omi. Simvastatin also inhibited mitochondrial fission with the loss of mitochondrial Drp1, an essential component of mitochondrial fission machinery. Thus, simvastatin activates novel apoptosis pathways in lung mesenchymal cells involving p53, IAP inhibitor release, and disruption of mitochondrial fission.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Cytochromes c/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung/pathology , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Serine Endopeptidases/metabolism , Simvastatin/pharmacology , Tumor Suppressor Protein p53/metabolism , Blotting, Western , Caspase 8/metabolism , Caspase 9/metabolism , Cholesterol/metabolism , Fibroblasts/drug effects , High-Temperature Requirement A Serine Peptidase 2 , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lung/metabolism , Mesoderm/cytology , Mitochondria/drug effects , Mitochondria/metabolism , Muscle, Smooth, Vascular/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , bcl-2-Associated X Protein/metabolism
6.
Cell Res ; 20(3): 314-31, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19935772

ABSTRACT

The complex formed by two members of the S100 calcium-binding protein family, S100A8/A9, exerts apoptosis-inducing activity in various cells of different origins. Here, we present evidence that the underlying molecular mechanisms involve both programmed cell death I (PCD I, apoptosis) and PCD II (autophagy)-like death. Treatment of cells with S100A8/A9 caused the increase of Beclin-1 expression as well as Atg12-Atg5 formation. S100A8/A9-induced cell death was partially inhibited by the specific PI3-kinase class III inhibitor, 3-methyladenine (3-MA), and by the vacuole H(+)-ATPase inhibitor, bafilomycin-A1 (Baf-A1). S100A8/A9 provoked the translocation of BNIP3, a BH3 only pro-apoptotic Bcl2 family member, to mitochondria. Consistent with this finding, DeltaTM-BNIP3 overexpression partially inhibited S100A8/A9-induced cell death, decreased reactive oxygen species (ROS) generation, and partially protected against the decrease in mitochondrial transmembrane potential in S100A8/A9-treated cells. In addition, either DeltaTM-BNIP3 overexpression or N-acetyl-L-cysteine co-treatment decreased lysosomal activation in cells treated with S100A8/A9. Our data indicate that S100A8/A9-promoted cell death occurs through the cross-talk of mitochondria and lysosomes via ROS and the process involves BNIP3.


Subject(s)
Apoptosis , Autophagy , Calgranulin A/metabolism , Calgranulin B/metabolism , Lysosomes/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Cell Line , Humans , Macrolides/pharmacology , Microtubule-Associated Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Small Ubiquitin-Related Modifier Proteins/metabolism , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/metabolism
7.
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
8.
Am J Respir Cell Mol Biol ; 37(6): 668-80, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17641293

ABSTRACT

Contractile airway smooth muscle (ASM) cells retain the ability for phenotype plasticity in response to multiple stimuli, which equips them with capacity to direct modeling and remodeling during development, and in disease states such as asthma. We have shown that endogenously expressed laminin is required for maturation of human ASM cells to a contractile phenotype, as occurs during ASM thickening in asthma. In this study, we profiled the expression of laminin-binding integrins alpha3beta1, alpha6beta1, and alpha7beta1, and tested whether they are required for laminin-induced myocyte maturation. Immunoblotting revealed that myocyte maturation induced by prolonged serum withdrawal, which was marked by the accumulation of contractile phenotype marker protein desmin, was also associated with the accumulation of alpha3A, alpha6A, and alpha7B. Flow cytometry revealed that alpha7B expression was a distinct feature of individual myocytes that acquired a contractile phenotype. siRNA knockdown of alpha7, but not alpha3 or alpha6, suppressed myocyte maturation. Thus, alpha7B is a novel marker of the contractile phenotype, and alpha7 expression is essential for human ASM cell maturation, which is a laminin-dependent process. These observations provide new insight into mechanisms that likely underpin normal development and remodeling associated with airways disease.


Subject(s)
Antigens, CD/metabolism , Integrin alpha Chains/metabolism , Laminin/metabolism , Muscle Contraction , Myocytes, Smooth Muscle/metabolism , Respiratory System/cytology , Respiratory System/metabolism , Antigens, CD/genetics , Biomarkers/metabolism , Cell Differentiation , Cell Line , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Integrin alpha Chains/genetics , Integrin alpha3/metabolism , Integrin alpha6/metabolism , Phenotype , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
9.
Am J Physiol Cell Physiol ; 293(1): C429-39, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17459944

ABSTRACT

We have previously demonstrated that long-term exposure of bovine tracheal smooth muscle (BTSM) strips to insulin induces a functional hypercontractile phenotype. To elucidate molecular mechanisms by which insulin might induce maturation of contractile phenotype airway smooth muscle (ASM) cells, we investigated effects of insulin stimulation in serum-free primary BTSM cell cultures on protein accumulation of specific contractile phenotypic markers and on the abundance and stability of mRNA encoding these markers. In addition, we used microscopy to assess insulin effects on ASM cell morphology, phenotype, and induction of phosphatidylinositol (PI) 3-kinase signaling. It was demonstrated that protein and mRNA levels of smooth muscle-specific contractile phenotypic markers, including sm-myosin, are significantly increased after stimulation of cultured BTSM cells with insulin (1 microM) for 8 days compared with cells treated with serum-free media, whereas mRNA stability was unaffected. In addition, insulin treatment promoted the formation of large, elongate ASM cells, characterized by dramatic accumulation of contractile phenotype marker proteins and phosphorylated p70(S6K) (downstream target of PI 3-kinase associated with ASM maturation). Insulin effects on protein accumulation and cell morphology were abrogated by combined pretreatment with the Rho kinase inhibitor Y-27632 (1 microM) or the PI 3-kinase inhibitor LY-294002 (10 microM), indicating that insulin increases the expression of contractile phenotypic markers in BTSM in a Rho kinase- and PI 3-kinase-dependent fashion. In conclusion, insulin increases transcription and protein expression of contractile phenotypic markers in ASM. This could have important implications for the use of recently approved aerosolized insulin formulations in diabetes mellitus.


Subject(s)
Contractile Proteins/metabolism , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Signal Transduction/drug effects , Trachea/drug effects , Amides/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Cattle , Cell Shape/drug effects , Cells, Cultured , Chromones/pharmacology , Contractile Proteins/genetics , Hypoglycemic Agents/metabolism , Insulin/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Microfilament Proteins/metabolism , Morpholines/pharmacology , Muscle Contraction/genetics , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Organ Culture Techniques , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Biosynthesis/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Receptor, IGF Type 2/metabolism , Receptor, Insulin/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Smooth Muscle Myosins/metabolism , Time Factors , Trachea/cytology , Trachea/metabolism , Transcription, Genetic/drug effects , rho-Associated Kinases , Calponins
10.
Respir Res ; 7: 117, 2006 Sep 12.
Article in English | MEDLINE | ID: mdl-16968549

ABSTRACT

BACKGROUND: Airway smooth muscle (ASM) contraction underlies acute bronchospasm in asthma. ASM cells can switch between a synthetic-proliferative phenotype and a contractile phenotype. While the effects of extracellular matrix (ECM) components on modulation of ASM cells to a synthetic phenotype have been reported, the role of ECM components on maturation of ASM cells to a contractile phenotype in adult lung is unclear. As both changes in ECM components and accumulation of contractile ASM are features of airway wall remodelling in asthma, we examined the role of the ECM protein, laminin, in the maturation of contractile phenotype in human ASM cells. METHODS: Human ASM cells were made senescence-resistant by stable expression of human telomerase reverse transcriptase. Maturation to a contractile phenotype was induced by 7-day serum deprivation, as assessed by immunoblotting for desmin and calponin. The role of laminin on ASM maturation was investigated by comparing the effects of exogenous laminin coated on culture plates, and of soluble laminin peptide competitors. Endogenous expression of laminin chains during ASM maturation was also measured. RESULTS: Myocyte binding to endogenously expressed laminin was required for ASM phenotype maturation, as laminin competing peptides (YIGSR or GRGDSP) significantly reduced desmin and calponin protein accumulation that otherwise occurs with prolonged serum deprivation. Coating of plastic cell culture dishes with different purified laminin preparations was not sufficient to further promote accumulation of desmin or calponin during 7-day serum deprivation. Expression of alpha2, beta1 and gamma1 laminin chains by ASM cells was specifically up-regulated during myocyte maturation, suggesting a key role for laminin-2 in the development of the contractile phenotype. CONCLUSION: While earlier reports suggest exogenously applied laminin slows the spontaneous modulation of ASM to a synthetic phenotype, we show for the first time that endogenously expressed laminin is required for ASM maturation to the contractile phenotype. As endogenously expressed laminin chains alpha2, beta1 and gamma1 are uniquely increased during myocyte maturation, these laminin chains may be key in this process. Thus, human ASM maturation appears to involve regulated endogenous expression of a select set of laminin chains that are essential for accumulation of contractile phenotype myocytes.


Subject(s)
Cell Enlargement , Laminin/biosynthesis , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Adult , Cell Line, Transformed , Cells, Cultured , Humans , Laminin/antagonists & inhibitors , Laminin/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism
11.
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
12.
Am J Respir Cell Mol Biol ; 31(1): 54-61, 2004 Jul.
Article in English | MEDLINE | ID: mdl-14975939

ABSTRACT

Mechanical strain affects airway myocyte phenotype, cytoskeletal architecture, proliferation, and contractile function. We hypothesized that (i) short-term mechanical strain modulates transcription of smooth muscle-specific gene promoters for SM22 and smooth muscle myosin heavy chain (smMHC); and (ii) strain-induced change is mediated by altered actin polymerization in association with activation of protein kinase C (PKC). Primary cultured canine tracheal myocytes were transiently transfected with luciferase reporter plasmids harboring a murine SM22, human smMHC, or artificial serum response factor (SRF)-specific gene promoter and then subjected to cyclic strain for 48 h. This strain protocol significantly reduced transcriptional activity of SM22 and smMHC promoters and an artificial SRF-dependent promoter by 55 +/- 5.9%, 57 +/- 6.4%, and 75 +/- 7.9%, respectively, with concomitant reduction in F/G actin ratio by 31 +/- 8%. PKC inhibitors, GF109203X or Gö6976, significantly attenuated these affects. Similar to strain, strain-independent activation of PKC inhibited SM22, smMHC, and SRF-dependent promoter activity by 61 +/- 4%, 66 +/- 5%, and 28 +/- 15%, respectively, and reduced the F/G actin ratio by 30 +/- 5%. Gel shift assay revealed that PKC activation led to decreased binding of the required transcription factor, SRF, to CArG elements in the SM22 promoter. These data suggest a previously unknown role for PKC isoforms in mechanosensitive signaling in airway myocytes that is associated with coordinated regulation of actin cytoskeletal dynamics and smooth muscle-specific gene transcription.


Subject(s)
Bronchi/enzymology , Gene Expression Regulation, Developmental/genetics , Microfilament Proteins/biosynthesis , Muscle Proteins/biosynthesis , Myocytes, Smooth Muscle/enzymology , Protein Kinase C/metabolism , Actins/biosynthesis , Animals , Bronchi/cytology , Cell Differentiation/genetics , Cells, Cultured , Dogs , Down-Regulation/genetics , Enzyme Inhibitors/pharmacology , Genes, Regulator/genetics , Microfilament Proteins/genetics , Muscle Proteins/genetics , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Promoter Regions, Genetic/genetics , Respiration/genetics , Serum Response Factor/biosynthesis , Serum Response Factor/genetics , Stress, Mechanical , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...