Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 12272, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37507487

ABSTRACT

Microtubule dysfunction has been implicated as a mediator of inflammation in multiple diseases such as disorders of the cardiovascular and neurologic systems. Tubulin polymerization promoting protein (Tppp) facilitates microtubule elongation and regulates tubulin acetylation through inhibition of cytosolic deacetylase enzymes. Pathologic alterations in microtubule structure and dynamics have been described in cystic fibrosis (CF) and associated with inflammation, however the causality and mechanism remain unclear. Likewise, Tppp has been identified as a potential modifier of CF airway disease severity. Here we directly assess the impact of microtubule dysfunction on infection and inflammation by interrogating wild type and a Tppp knockout mouse model (Tppp - / -). Mice are challenged with a clinical isolate of Pseudomonas aeruginosa-laden agarose beads and assessed for bacterial clearance and inflammatory markers. Tppp - / - mouse model demonstrate impaired bacterial clearance and an elevated inflammatory response compared to control mice. These data are consistent with the hypothesis microtubule dysregulation is sufficient to lead to CF-like airway responses in mice.


Subject(s)
Cystic Fibrosis , Nerve Tissue Proteins , Tubulin , Animals , Mice , Cystic Fibrosis/metabolism , Microtubules/metabolism , Nerve Tissue Proteins/metabolism , Polymerization , Tubulin/metabolism
2.
Am J Physiol Lung Cell Mol Physiol ; 322(3): L333-L347, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34986321

ABSTRACT

Several aspects of the cell biology of cystic fibrosis (CF) epithelial cells are altered including impaired lipid regulation, disrupted intracellular transport, and impaired microtubule regulation. It is unclear how the loss of cystic fibrosis transmembrane conductance regulator (CFTR) function leads to these differences. It is hypothesized that the loss of CFTR function leads to altered regulation of carbonic anhydrase (CA) activity resulting in cellular phenotypic changes. In this study, it is demonstrated that CA2 protein expression is reduced in CF model cells, primary mouse nasal epithelial (MNE) cells, excised MNE tissue, and primary human nasal epithelial cells (P < 0.05). This corresponds to a decrease in CA2 RNA expression measured by qPCR as well as an overall reduction in CA activity in primary CF MNEs. The addition of CFTR-inhibitor-172 to WT MNE cells for ≥24 h mimics the significantly lower protein expression of CA2 in CF cells. Treatment of CF cells with l-phenylalanine (L-Phe), an activator of CA activity, restores endosomal transport through an effect on microtubule regulation in a manner dependent on soluble adenylate cyclase (sAC). This effect can be blocked with the CA2-selective inhibitor dorzolamide. These data suggest that the loss of CFTR function leads to the decreased expression of CA2 resulting in the downstream cell signaling alterations observed in CF.


Subject(s)
Carbonic Anhydrases , Cystic Fibrosis , Adenylyl Cyclases/metabolism , Animals , Carbonic Anhydrases/metabolism , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/metabolism , Mice , Phenotype
3.
Sci Rep ; 10(1): 16278, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33004910

ABSTRACT

Cystic fibrosis (CF) patients experience heightened levels of anxiety and depression. Stress from dealing with chronic disease and rigorous treatment regimens certainly are primary contributors to these outcomes. We previously have demonstrated that microtubule alterations in CF are linked to a number of CF phenotypes including growth regulation and inflammatory responses to airway bacterial challenge. Deletion of histone deactelyase 6 (HDAC6), a cytosolic deacetylase that regulates tubulin acetylation, in CF mice restores growth and inflammatory phenotypes to wild type (WT) profiles. In this study, the hypothesis that Hdac6 depletion in CF mice would impact behaviors since Hda6 inhibition has been previously reported to have anti-depressive properties. Data demonstrate that CF mice exhibit reduced activity and reduced open arm time in an elevated plus maze test which can be consistent with anxiety-like behavior. CF mice also exhibit depression-like behaviors compared to WT mice in an age dependent manner. By eight weeks of age, CF mice exhibit significantly more immobile time in the tail-suspension test, however, Hdac6 depletion reverses the depressive phenotype. These data demonstrate that loss of CFTR function may predispose patients to experience depression and that this behavior is Hdac6 dependent.


Subject(s)
Cystic Fibrosis/complications , Depression/etiology , Histone Deacetylase 6/physiology , Animals , Anxiety/etiology , Cystic Fibrosis/psychology , Disease Models, Animal , Female , Male , Maze Learning , Mice , Mice, Knockout
4.
Am J Physiol Lung Cell Mol Physiol ; 318(6): L1145-L1157, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32267731

ABSTRACT

We have demonstrated previously that intracellular transport is impaired in cystic fibrosis (CF) epithelial cells. This impairment is related to both growth and inflammatory regulation in CF cell and animal models. Understanding how transport in CF cells is regulated and identifying means to manipulate that regulation are key to identifying new therapies that can address key CF phenotypes. It was hypothesized that resveratrol could replicate these benefits since it interfaces with multiple pathways identified to affect microtubule regulation in CF. It was found that resveratrol treatment significantly restored intracellular transport as determined by monitoring both cholesterol distribution and the distribution of rab7-positive organelles in CF cells. This restoration of intracellular transport is due to correction of both microtubule formation rates and microtubule acetylation in cultured CF cell models and primary nasal epithelial cells. Mechanistically, the effect of resveratrol on microtubule regulation and intracellular transport was dependent on peroxisome proliferator-activated receptor-γ signaling and its ability to act as a pan-histone deacetylase (HDAC) inhibitor. Resveratrol represents a candidate compound with known anti-inflammatory properties that can restore both microtubule formation and acetylation in CF epithelial cells.


Subject(s)
Cystic Fibrosis/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Intracellular Space/metabolism , Resveratrol/pharmacology , 1-Methyl-3-isobutylxanthine/pharmacology , Acetylation/drug effects , Biological Transport/drug effects , Carbazoles/pharmacology , Cells, Cultured , Cholesterol/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Humans , Intracellular Space/drug effects , Microtubules/drug effects , Microtubules/metabolism , Nose/pathology , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , Phosphodiesterase Inhibitors/pharmacology , Resorcinols/pharmacology , Signal Transduction/drug effects , Sirtuins/metabolism , Stilbenes/pharmacology , Tubulin/metabolism
5.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L1081-L1093, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30892081

ABSTRACT

The use of high-dose ibuprofen as an anti-inflammatory therapy in cystic fibrosis (CF) has been shown to be an effective intervention although use is limited due to potential adverse events. Identifying the mechanism of ibuprofen efficacy would aid in the development of new therapies that avoid these adverse events. Previous findings demonstrated that ibuprofen treatment restores the regulation of microtubule dynamics in CF epithelial cells through a 5'-adenosine monophosphate-activated protein kinase (AMPK)-dependent mechanism. The goal of this study is to define the AMPK pathway that leads to microtubule regulation. Here, it is identified that inhibition of acetyl-CoA carboxylase (ACC) is the key step in mediating the AMPK effect. ACC inhibition with 5-(tetradecyloxy)-2-furoic acid (TOFA) increases microtubule reformation rates in cultured and primary CF epithelial cells to wild-type (WT) rates. TOFA treatment also restores microtubule-dependent distribution of cholesterol and Rab7-positive organelles, as well as reduces expression of the proinflammatory signaling molecule RhoA to WT levels. ACC activation with citrate replicates these CF phenotypes in WT cells further supporting the role of AMPK signaling through ACC as a key mediator in CF cell signaling. It is concluded that ACC inhibition is the key step in the efficacy of AMPK activation at the cellular level and could represent a novel site of therapeutic intervention to address inflammation in CF.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acetyl-CoA Carboxylase/antagonists & inhibitors , Cystic Fibrosis/pathology , Epithelial Cells/metabolism , Microtubules/pathology , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Child , Cholesterol/metabolism , Female , Furans/pharmacology , Humans , Ibuprofen/pharmacology , Male , Mice, Knockout , Sf9 Cells , Spodoptera , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins , rhoA GTP-Binding Protein/biosynthesis
6.
Sci Rep ; 7(1): 3676, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28623308

ABSTRACT

Growth failure in cystic fibrosis (CF) patients has been well-documented and shown to correlate with poorer disease outcomes. This observation is also true in CF animal models, including mouse, pig, rat, and ferret. The etiology underlying growth deficits is unknown, and our previous work demonstrated reduced tubulin acetylation in CF cell models and tissue that is correctable by inhibition of histone deacetylase-6 (HDAC6). Here, we hypothesize that loss of HDAC6 will improve growth phenotype in a CF mouse model. Hdac6 knockout mice were crossed with F508del (CF) mice to generate F508del/Hdac6 (CF/HDA) mice. Growth, fat deposits, survival, and bioelectric measurements were analyzed. CF/HDA mice displayed improvements in length and weight with no correction of CFTR function. Mechanistically, Igf1 levels likely account for increased length and improvements in fertility. Weight gain is attributed to increased fat deposits potentially mediated by increased adipocyte differentiation. CF-related growth deficits can be improved via inhibition of HDAC6, further implicating it as a potential therapeutic target for CF.


Subject(s)
Cystic Fibrosis/genetics , Genetic Predisposition to Disease , Growth Disorders/diagnosis , Histone Deacetylase 6/deficiency , Adiposity , Animals , Biomarkers , Cystic Fibrosis/complications , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Models, Animal , Female , Fluorescent Antibody Technique , Growth Disorders/etiology , Growth Disorders/metabolism , Histone Deacetylase 6/genetics , Histone Deacetylase 6/metabolism , Male , Mice , Mice, Knockout
7.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L317-27, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27317686

ABSTRACT

High-dose ibuprofen, an effective anti-inflammatory therapy for the treatment of cystic fibrosis (CF), has been shown to preserve lung function in a pediatric population. Despite its efficacy, few patients receive ibuprofen treatment due to potential renal and gastrointestinal toxicity. The mechanism of ibuprofen efficacy is also unclear. We have previously demonstrated that CF microtubules are slower to reform after depolymerization compared with respective wild-type controls. Slower microtubule dynamics in CF cells are responsible for impaired intracellular transport and are related to inflammatory signaling. Here, it is identified that high-dose ibuprofen treatment in both CF cell models and primary CF nasal epithelial cells restores microtubule reformation rates to wild-type levels, as well as induce extension of microtubules to the cell periphery. Ibuprofen treatment also restores microtubule-dependent intracellular transport monitored by measuring intracellular cholesterol transport. These effects are specific to ibuprofen as other cyclooxygenase inhibitors have no effect on these measures. Effects of ibuprofen are mimicked by stimulation of AMPK and blocked by the AMPK inhibitor compound C. We conclude that high-dose ibuprofen treatment enhances microtubule formation in CF cells likely through an AMPK-related pathway. These findings define a potential mechanism to explain the efficacy of ibuprofen therapy in CF.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Cystic Fibrosis/drug therapy , Epithelial Cells/metabolism , Ibuprofen/pharmacology , Microtubules/metabolism , Acetylation , Adenylate Kinase , Cell Line , Cystic Fibrosis/pathology , Drug Evaluation, Preclinical , Enzyme Activation , Epithelial Cells/drug effects , Guanine Nucleotide Exchange Factors/metabolism , Humans , Primary Cell Culture , Protein Multimerization , Protein Processing, Post-Translational , Shelterin Complex , Telomere-Binding Proteins/metabolism
8.
Am J Respir Cell Mol Biol ; 53(6): 853-62, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25955407

ABSTRACT

The regulation of microtubule dynamics in cystic fibrosis (CF) epithelial cells and the consequences of reduced rates of microtubule polymerization on downstream CF cellular events, such as cholesterol accumulation, a marker of impaired intracellular transport, are explored here. It is identified that microtubules in both CF cell models and in primary CF nasal epithelial cells repolymerize at a slower rate compared with respective controls. Previous studies suggest a role for cAMP in modulating organelle transport in CF cells, implicating a role for exchange protein activated by cAMP (EPAC) 1, a regulator of microtubule elongation, as a potential mechanism. EPAC1 activity is reduced in CF cell models and in Cftr(-/-) mouse lung compared with respective non-CF controls. Stimulation of EPAC1 activity with the selective EPAC1 agonist, 8-cpt-2-O-Me-cAMP, stimulates microtubule repolymerization to wild-type rates in CF cells. EPAC1 activation also alleviates cholesterol accumulation in CF cells, suggesting a direct link between microtubule regulation and intracellular transport. To verify the relationship between transport and microtubule regulation, expression of the protein, tubulin polymerization-promoting protein, was knocked down in non-CF human tracheal (9/HTEo(-)) cells to mimic the microtubule dysregulation in CF cells. Transduced cells with short hairpin RNA targeting tubulin polymerization-promoting protein exhibit CF-like perinuclear cholesterol accumulation and other cellular manifestations of CF cells, thus supporting a role for microtubule regulation as a mechanism linking CFTR function to downstream cellular manifestation.


Subject(s)
Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Guanine Nucleotide Exchange Factors/physiology , Microtubules/metabolism , Animals , Cell Line , Cyclic AMP , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Humans , Kinetics , Mice, Knockout , Microtubules/pathology , Protein Multimerization , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology
9.
Am J Physiol Lung Cell Mol Physiol ; 305(6): L419-31, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23873844

ABSTRACT

Dysfunctional cystic fibrosis transmembrane conductance regulator (CFTR) leads to many cellular consequences, including perinuclear accumulation of free cholesterol due to impaired endosomal transport. The hypothesis being tested is that CF-related perinuclear cholesterol accumulation due to disrupted endocytic trafficking occurs as a result of reduced microtubule (MT) acetylation. Here, it is identified that acetylated-α-tubulin (Ac-tub) content is reduced by ∼40% compared with respective wild-type controls in both cultured CF cell models (IB3) and primary Cftr-/- mouse nasal epithelial tissue. Histone deacetylase 6 (HDAC6) has been shown to regulate MT acetylation, which provides reasonable grounds to test its impact on reduced Ac-tub content on CF cellular phenotypes. Inhibition of HDAC6, either through tubastatin treatment or HDAC6 knockdown in CF cells, increases Ac-tub content and results in redistributed free cholesterol and reduced stimulation of NF-κB activity. Mechanistically, endoplasmic reticulum stress, which is widely reported in CF and leads to aggresome formation, is identified as a regulator of MT acetylation. F508del CFTR correction with C18 in primary airway epithelial cells restores MT acetylation and cholesterol transport. A significant role for phosphatidyl inositol-3 kinase p110α is also identified as a regulator of MT acetylation.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Cystic Fibrosis/physiopathology , Epithelial Cells/metabolism , Microtubules/metabolism , Acetylation , Animals , Cholesterol/metabolism , Class I Phosphatidylinositol 3-Kinases , Endoplasmic Reticulum Stress/physiology , Endosomes/physiology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Mice , NF-kappa B/metabolism , Nasal Mucosa/metabolism , Phosphoinositide-3 Kinase Inhibitors , Tubulin/metabolism
10.
J Lipid Res ; 53(7): 1268-76, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22523395

ABSTRACT

Cystic fibrosis (CF) cells exhibit an increase in the protein expression of ß-arrestin-2 (ßarr2) coincident with perinuclear accumulation of free cholesterol. Arrestins are proteins that both serve as broad signaling regulators and contribute to G-protein coupled receptor internalization after agonist stimulation. The hypothesis of this study is that ßarr2 is an important component in the mechanisms leading to cholesterol accumulation characteristic of CF cells. To test this hypothesis, epithelial cells stably expressing GFP-tagged ßarr2 (ßarr2-GFP) and respective GFP-expressing control cells (cont-GFP) were analyzed by filipin staining. The ßarr2-GFP cells show a late endosomal/lysosomal cholesterol accumulation that is identical to that seen in CF cells. This ßarr2-mediated accumulation is sensitive to Rp-cAMPS treatment, and depleting ßarr2 expression in CF-model cells by shRNA alleviates cholesterol accumulation compared with controls. Cftr/ßarr2 double knockout mice also exhibit wild-type (WT) levels of cholesterol synthesis, and WT profiles of signaling protein expression have previously been shown to be altered in CF due to cholesterol-related pathways. These data indicate a significant regulatory role for ßarr2 in the development of CF-like cholesterol accumulation and give further insight into cholesterol processing mechanisms. An impact of ßarr2 expression on Niemann-Pick type C-1 (NPC1)-containing organelle movement is proposed as the mechanism of ßarr2-mediated alterations on cholesterol processing. It is concluded that ßarr2 expression contributes to altered cholesterol trafficking observed in CF cells.


Subject(s)
Arrestins/metabolism , Cholesterol/metabolism , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Animals , Arrestins/deficiency , Arrestins/genetics , Cells, Cultured , Cholesterol/biosynthesis , Cholesterol/genetics , Cystic Fibrosis/genetics , Epithelial Cells/pathology , Humans , Mice , Mice, Inbred CFTR , Mice, Knockout , Phenotype , beta-Arrestin 2 , beta-Arrestins
11.
Biochemistry ; 50(27): 6022-9, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21644508

ABSTRACT

Previous work demonstrated that cystic fibrosis (CF) cells exhibit an increase in cAMP-mediated signaling as a characteristic response to lost CFTR function. Evidence for increased cAMP-mediated signaling in CF included increased phosphorylation of the cAMP response element binding protein (CREB) and elevated ß-arrestin-2 (ßarr2) expression. However, subsequent studies reveal that CREB activation in CF cells is independent of protein kinase-A (PKA). The goal of this study is to test the hypothesis that elevated ßarr2 expression leads to increased CREB activation in a PKA-independent mechanism. ßarr2-GFP expressing tracheal epithelial cells (ßarr2-GFP) exhibit an increase of pCREB content and subsequent CRE activation compared to GFP expressing control cells. ßarr2 activation of the ERK cascade represents a candidate mechanism leading to CREB activation. ERK exhibits increased activation in ßarr2-GFP cells compared to cont-GFP cells, and ERK inhibition diminishes CRE activation in both GFP and ßarr2-GFP cells. To test directly whether CREB regulation in CF is ßarr2-dependent, nasal epithelium excised from wt mice (Cftr +/+; ßarr2 +/+), CF mice (Cftr -/-; ßarr2 +/+), and DKO mice (Cftr -/-; ßarr2 -/-) were analyzed for pCREB protein content. Removal of ßarr2 expression from CF mice reduces both pCREB and pERK content to wt levels. These data indicate that CF-related CREB regulation is mediated directly through ßarr2 expression via the ERK pathway.


Subject(s)
Arrestins/physiology , Cyclic AMP Response Element-Binding Protein/metabolism , Cystic Fibrosis/metabolism , Signal Transduction , Animals , Arrestins/biosynthesis , Arrestins/deficiency , Cell Line , Cells, Cultured , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Disease Models, Animal , Gene Expression Regulation/physiology , Humans , MAP Kinase Signaling System/genetics , Mice , Mice, Inbred CFTR , Mice, Knockout , Mice, Transgenic , Phenotype , Phosphorylation/genetics , Protein Structure, Tertiary/genetics , Signal Transduction/genetics , beta-Arrestin 2 , beta-Arrestins
12.
Am J Physiol Lung Cell Mol Physiol ; 295(5): L809-19, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18790990

ABSTRACT

The goal of this study was to identify a mechanism regulating cholesterol accumulation in cystic fibrosis (CF) cells. Both CFTR activation and expression are regulated by the cAMP pathway, and it is hypothesized that a feedback response involving this pathway may be involved in the phenotype of cholesterol accumulation. To examine the role of the cAMP pathway in cholesterol accumulation, we treated two CF model cell lines with the Rp diastereomer of adenosine 3',5'-cyclic monophosphorothioate (Rp-cAMPS) and visualized by filipin staining. Rp-cAMPS treatment eliminated cholesterol accumulation in CF cells, whereas 8-bromo-cAMP treatment led to cholesterol accumulation in wild-type cells. To confirm these findings in an independent model system, we also examined the role of cAMP in modulating cholesterol accumulation in Niemann-Pick type C (NPC) fibroblasts. Expression of the protein related to NPC, NPC1, is also directly regulated by cAMP; therefore, it is postulated that NPC cells exhibit the same cAMP-mediated control of cholesterol accumulation. Cholesterol accumulation in NPC cells also was reduced by the presence of Rp-cAMPS. Expression of beta-arrestin-2 (betaarr2), a marker of cellular response to cAMP signaling, was significantly elevated in CF model cells, Cftr(-/-) MNE, primary tissue obtained by nasal scrapes from CF subjects, and in NPC fibroblasts compared with respective controls.


Subject(s)
Cholesterol/metabolism , Cyclic AMP/pharmacology , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Arrestins/metabolism , Cell Line , Cyclic AMP/analogs & derivatives , Cyclic AMP Response Element-Binding Protein/metabolism , Epithelium/drug effects , Epithelium/metabolism , Epithelium/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Phosphorylation/drug effects , Protein Transport/drug effects , Receptors, Adrenergic, beta-2/metabolism , Thionucleotides/pharmacology , beta-Arrestin 2 , beta-Arrestins
13.
Biochim Biophys Acta ; 1772(7): 748-54, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17499483

ABSTRACT

Niemann-Pick type C (NPC) disease is characterized at the cellular level by the intracellular accumulation of free cholesterol. We have previously identified a similar phenotype in cystic fibrosis (CF) cell models that results in the activation of the small GTPase RhoA. The hypothesis of this study was that NPC cells would also exhibit an increase in small GTPase activation. An examination of the active, GTP-bound form of GTPases revealed a basal increase in the content of the active-form Ras and RhoA small GTPases in NPC fibroblasts compared to wt controls. To assess whether this increase in GTP-bound Ras and RhoA manifests a functional outcome, the expression of the proliferation control proteins p21/waf1 and cyclin D were examined. Consistent with increased GTPase signaling, p21/waf1 expression is reduced and cyclin D expression is elevated in NPC fibroblasts. Interestingly, cell growth rate is not altered in NPC fibroblasts compared to wt cells. However, NPC sensitivity to statin treatment is reversed by addition of the isoprenoid geranylgeranyl pyrophosphate (GGPP), a modifier of RhoA. It is concluded that Ras and RhoA basal activation is elevated in NPC fibroblasts and has an impact on cell survival pathways.


Subject(s)
Cell Proliferation , GTP Phosphohydrolases/metabolism , Niemann-Pick Disease, Type C/pathology , Signal Transduction , Blotting, Western , Cells, Cultured , Enzyme Activation , Fibroblasts/metabolism , Humans , Niemann-Pick Disease, Type C/enzymology
14.
Cell Signal ; 19(5): 923-31, 2007 May.
Article in English | MEDLINE | ID: mdl-17197157

ABSTRACT

SMAD3 is a transcription factor that mediates TGF-beta1 signaling and is known to be important in many of the cellular processes that regulate fibrosis and inflammation. Although several studies have examined SMAD3 activation, little is known about the control of SMAD3 expression. It is well established that the mitogen-activated protein kinase (MAPK) pathway is responsive to TGF-beta1 stimulation and coordinates with SMAD signaling in many cases; therefore, the hypothesis of this study is that the MAPK pathway will be involved in the regulation of SMAD3 expression. Using a SMAD3 promoter construct, we demonstrate that inhibition of either c-Jun-N-terminal kinase (JNK) or p38 activity has little effect on SMAD3 promoter function. Inhibition of mitogen-activated protein kinase kinase-1 (MEK1) with either PD98059 or UO126, however, results in a substantial dose-dependent inhibition of SMAD3 promoter activity. Further studies confirm that promoter activity correlates with protein expression by demonstrating reduced SMAD3 protein expression in A549 cells and airway smooth muscle cells after treatment with MEK1 inhibitors. Positive regulation of SMAD3 expression is also demonstrated by expression of a constitutively active (ca)-MEK1 construct, where the presence of ca-MEK1 resulted in increased SMAD3 protein expression. These data lead to the conclusion that MEK1 is an important regulator of SMAD3 expression.


Subject(s)
Gene Expression Regulation , MAP Kinase Kinase 1/metabolism , Muscle, Smooth/metabolism , Respiratory Mucosa/metabolism , Smad3 Protein/genetics , Animals , Base Sequence , Cell Line, Tumor , Cells, Cultured , Conserved Sequence , DNA , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Rats , Respiratory System/cytology , Sequence Alignment , Signal Transduction , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism , Species Specificity , Transforming Growth Factor beta1/metabolism
15.
Am J Respir Cell Mol Biol ; 31(5): 538-43, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15256387

ABSTRACT

Recent data demonstrate that inhibition of 3-hydroxy-3-methylglutaryl-CoA reductase restores normal signal transducer and activator of transcription-1 and inducible nitric oxide synthase expression regulation in cystic fibrosis (CF) cells through the modulation of RhoA function. These findings lead to the hypothesis that alterations in the cholesterol synthesis pathway may be an initiating factor in CF-related cell signaling regulation. A disease with a known lesion in the cholesterol synthesis pathway is Niemann-Pick type C (NPC). The hypothesis of this study is that CF cells and NPC fibroblasts share a common mechanistic lesion and should exhibit similar cell signaling alterations. NPC fibroblasts exhibit similar alterations in signal transducer and activator of transcription-1, RhoA, SMAD3, and nitric oxide synthase protein expression that characterize CF. Further comparison reveals NPC-like accumulation of free cholesterol in two cultured models of CF epithelial cells. These data identify novel signaling changes in NPC, demonstrate the cholesterol-synthesis pathway is a likely source of CF-related cell signaling changes, and that cultured CF cells exhibit impaired cholesterol processing.


Subject(s)
Cystic Fibrosis/enzymology , Niemann-Pick Diseases/enzymology , Actins/metabolism , Animals , Blotting, Western , Cells, Cultured , Cholesterol/metabolism , Disease Models, Animal , Epithelial Cells/cytology , Fibroblasts/metabolism , Filipin/chemistry , Filipin/metabolism , Humans , Mice , Mice, Transgenic , Mutation, Missense , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Phenotype , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Sphingomyelin Phosphodiesterase/metabolism , Temperature , rhoA GTP-Binding Protein/metabolism
16.
Am J Physiol Lung Cell Mol Physiol ; 285(6): L1286-95, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12948935

ABSTRACT

The expression of the inducible form of nitric oxide synthase (NOS2) is reduced in cystic fibrosis (CF) epithelium despite the presence of aggressive inflammation. A potential mechanism for reduced NOS2 expression in CF is diminished signal transducer and activator of transcription-1 (STAT1) activity, possibly due to an increase in expression of protein inhibitor of activated STAT1 (PIAS1). Previous evidence also suggests that NOS2 expression can be negatively regulated by increased activation of the GTPase RhoA, leading to the hypothesis that CF-related increases in PIAS1 expression and altered STAT1 signaling may be mediated by Rho GTPase function. Consistent with this hypothesis, data demonstrate increased expression of RhoA in two models of CF epithelium with a proportional increase in the active GTP-bound RhoA. Mouse embryonic fibroblasts null for p190B Rho GTPase-activating protein exhibit increased RhoA protein content and activation, similar to what is observed in CF models, and also exhibit CF-like alterations in STAT1 regulation, including decreased STAT1 activation, increased PIAS1 protein expression, and reduced NOS2 induction, implicating RhoA-mediated signaling in CF-related STAT1 alterations. Inhibition of the Rho GTPase pathway at the level of isoprenoid/cholesterol synthesis with mevastatin reduces PIAS1 expression, increases STAT1 activation, and restores NOS2 expression in models of CF epithelium, suggesting that pharmacological inhibition of the isoprenoid synthesis/Rho GTPase pathway may represent a potential avenue for therapeutic intervention for CF.


Subject(s)
Cystic Fibrosis/physiopathology , DNA-Binding Proteins/metabolism , Lovastatin/analogs & derivatives , Nitric Oxide Synthase/genetics , Respiratory Mucosa/physiopathology , Signal Transduction/physiology , Trans-Activators/metabolism , Adenocarcinoma, Bronchiolo-Alveolar , Anticholesteremic Agents/pharmacology , Cell Line, Tumor , Cholesterol/biosynthesis , Cystic Fibrosis/metabolism , Gene Expression Regulation, Enzymologic , Humans , Lovastatin/pharmacology , Lung Neoplasms , Nitric Oxide Synthase Type II , Protein Inhibitors of Activated STAT , Proteins/genetics , Proteins/metabolism , Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , STAT1 Transcription Factor , Signal Transduction/drug effects , Terpenes/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
17.
Am J Physiol Lung Cell Mol Physiol ; 283(3): L604-11, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12169580

ABSTRACT

The aberrant dysregulation of the inducible form of nitric oxide synthase (NOS2) is thought to play a role in many inflammatory disorders including cystic fibrosis (CF). The complex regulation of NOS2 expression is the subject of intense investigation, and one intriguing regulatory pathway known to influence NOS2 expression is the Rho GTPase cascade. We examined NOS2 regulation in response to inflammatory cytokines in a human alveolar epithelial cell line treated with inhibitors of different upstream and downstream components of the Rho GTPase pathway to better define potential signaling mechanisms. Statin-mediated 3-hydroxy-3-methylglutaryl-CoA reductase inhibition increased cytokine-dependent activation of the NOS2 promoter, reversible by the addition of geranylgeranyl pyrphosphate. However, inhibition of Rho-associated kinase (ROCK) with Y-27632 resulted in a decrease in NOS2 promoter activity, yet an increase in NOS2 mRNA and protein levels. Our results suggest that prenylation events influence NOS2 promoter activity independently of the Rho GTPase pathway and that Rho GTPase signaling mediated through ROCK suppresses NOS2 production downstream of promoter function at the message and protein level.


Subject(s)
Lovastatin/analogs & derivatives , Nitric Oxide Synthase/metabolism , Pulmonary Alveoli/enzymology , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , Amides/pharmacology , Dimethylallyltranstransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Epithelial Cells/enzymology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins , Lovastatin/pharmacology , NF-kappa B/metabolism , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Promoter Regions, Genetic/drug effects , Protein Serine-Threonine Kinases/pharmacology , Pulmonary Alveoli/cytology , Pyridines/pharmacology , RNA, Messenger/metabolism , Transcription Factor AP-1/metabolism , Tumor Cells, Cultured , rho-Associated Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...