1
|
Hernandez-Lara MA, Richard J, Deshpande DA. Diacylglycerol kinase is a keystone regulator of signaling relevant to the pathophysiology of asthma. Am J Physiol Lung Cell Mol Physiol 2024; 327:L3-L18. [PMID: 38742284 PMCID: PMC11380957 DOI: 10.1152/ajplung.00091.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/05/2024] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
Signal transduction by G protein-coupled receptors (GPCRs), receptor tyrosine kinases (RTKs) and immunoreceptors converge at the activation of phospholipase C (PLC) for the hydrolysis of phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG). This is a point for second-messenger bifurcation where DAG via protein kinase C (PKC) and IP3 via calcium activate distinct protein targets and regulate cellular functions. IP3 signaling is regulated by multiple calcium influx and efflux proteins involved in calcium homeostasis. A family of lipid kinases belonging to DAG kinases (DGKs) converts DAG to phosphatidic acid (PA), negatively regulating DAG signaling and pathophysiological functions. PA, through a series of biochemical reactions, is recycled to produce new molecules of PIP2. Therefore, DGKs act as a central switch in terminating DAG signaling and resynthesis of membrane phospholipids precursor. Interestingly, calcium and PKC regulate the activation of α and ζ isoforms of DGK that are predominantly expressed in airway and immune cells. Thus, DGK forms a feedback and feedforward control point and plays a crucial role in fine-tuning phospholipid stoichiometry, signaling, and functions. In this review, we discuss the previously underappreciated complex and intriguing DAG/DGK-driven mechanisms in regulating cellular functions associated with asthma, such as contraction and proliferation of airway smooth muscle (ASM) cells and inflammatory activation of immune cells. We highlight the benefits of manipulating DGK activity in mitigating salient features of asthma pathophysiology and shed light on DGK as a molecule of interest for heterogeneous diseases such as asthma.
Collapse
Affiliation(s)
- Miguel A Hernandez-Lara
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Joshua Richard
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| | - Deepak A Deshpande
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States
| |
Collapse
|
2
|
Chiba Y, Yamane Y, Sato T, Suto W, Hanazaki M, Sakai H. Extracellular acidification attenuates bronchial contraction via an autocrine activation of EP 2 receptor: Its diminishment in murine experimental asthma. Respir Physiol Neurobiol 2024; 324:104251. [PMID: 38492830 DOI: 10.1016/j.resp.2024.104251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/03/2024] [Accepted: 03/09/2024] [Indexed: 03/18/2024]
Abstract
PURPOSE Extracellular acidification is a major component of tissue inflammation, including airway inflammation in asthmatics. However, its physiological/pathophysiological significance in bronchial function is not fully understood. Currently, the functional role of extracellular acidification on bronchial contraction was explored. METHODS Left main bronchi were isolated from male BALB/c mice. Epithelium-removed tissues were exposed to acidic pH under submaximal contraction induced by 10-5 M acetylcholine in the presence or absence of a COX inhibitor indomethacin (10-6 M). Effects of AH6809 (10-6 M, an EP2 receptor antagonist), BW A868C (10-7 M, a DP receptor antagonist) and CAY10441 (3×10-6 M, an IP receptor antagonist) on the acidification-induced change in tension were determined. The release of prostaglandin E2 (PGE2) from epithelium-denuded tissues in response to acidic pH was assessed using an ELISA. RESULTS In the bronchi stimulated with acetylcholine, change in the extracellular pH from 7.4 to 6.8 caused a transient augmentation of contraction followed by a sustained relaxing response. The latter inhibitory response was abolished by indomethacin and AH6809 but not by BW A868C or CAY10441. Both indomethacin and AH6809 significantly increased potency and efficacy of acetylcholine at pH 6.8. Stimulation with low pH caused an increase in PGE2 release from epithelium-denuded bronchi. Interestingly, the acidic pH-induced bronchial relaxation was significantly reduced in a murine asthma model that had a bronchial hyperresponsiveness to acetylcholine. CONCLUSION Taken together, extracellular acidification could inhibit the bronchial contraction via autocrine activation of EP2 receptors. The diminished acidic pH-mediated inhibition of bronchial tone may contribute to excessive bronchoconstriction in inflamed airways such as asthma.
Collapse
Affiliation(s)
| | - Yamato Yamane
- Laboratory of Molecular Biology and Physiology, Japan
| | - Tsubasa Sato
- Laboratory of Molecular Biology and Physiology, Japan
| | - Wataru Suto
- Laboratory of Molecular Biology and Physiology, Japan
| | - Motohiko Hanazaki
- Department of Anesthesiology and Intensive Care Medicine, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Hiroyasu Sakai
- Laboratory of Biomolecular Pharmacology, Hoshi University School of Pharmacy, Tokyo, Japan
| |
Collapse
|
3
|
Hernandez-Lara MA, Yadav SK, Conaway S, Shah SD, Penn RB, Deshpande DA. Crosstalk between diacylglycerol kinase and protein kinase A in the regulation of airway smooth muscle cell proliferation. Respir Res 2023; 24:155. [PMID: 37301818 PMCID: PMC10257838 DOI: 10.1186/s12931-023-02465-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
BACKGROUND Diacylglycerol kinase (DGK) regulates intracellular signaling and functions by converting diacylglycerol (DAG) into phosphatidic acid. We previously demonstrated that DGK inhibition attenuates airway smooth muscle (ASM) cell proliferation, however, the mechanisms mediating this effect are not well established. Given the capacity of protein kinase A (PKA) to effect inhibition of ASM cells growth in response to mitogens, we employed multiple molecular and pharmacological approaches to examine the putative role of PKA in the inhibition of mitogen-induced ASM cell proliferation by the small molecular DGK inhibitor I (DGK I). METHODS We assayed cell proliferation using CyQUANT™ NF assay, protein expression and phosphorylation using immunoblotting, and prostaglandin E2 (PGE2) secretion by ELISA. ASM cells stably expressing GFP or PKI-GFP (PKA inhibitory peptide-GFP chimera) were stimulated with platelet-derived growth factor (PDGF), or PDGF + DGK I, and cell proliferation was assessed. RESULTS DGK inhibition reduced ASM cell proliferation in cells expressing GFP, but not in cells expressing PKI-GFP. DGK inhibition increased cyclooxygenase II (COXII) expression and PGE2 secretion over time to promote PKA activation as demonstrated by increased phosphorylation of (PKA substrates) VASP and CREB. COXII expression and PKA activation were significantly decreased in cells pre-treated with pan-PKC (Bis I), MEK (U0126), or ERK2 (Vx11e) inhibitors suggesting a role for PKC and ERK in the COXII-PGE2-mediated activation of PKA signaling by DGK inhibition. CONCLUSIONS Our study provides insight into the molecular pathway (DAG-PKC/ERK-COXII-PGE2-PKA) regulated by DGK in ASM cells and identifies DGK as a potential therapeutic target for mitigating ASM cell proliferation that contributes to airway remodeling in asthma.
Collapse
Affiliation(s)
- Miguel A. Hernandez-Lara
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Santosh Kumar Yadav
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Stanley Conaway
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Sushrut D. Shah
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Raymond B. Penn
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| | - Deepak A. Deshpande
- Department of Medicine, Center for Translational Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107 USA
| |
Collapse
|
4
|
Yadav SK, Sharma P, Shah SD, Panettieri RA, Kambayashi T, Penn RB, Deshpande DA. Autocrine regulation of airway smooth muscle contraction by diacylglycerol kinase. J Cell Physiol 2022; 237:603-616. [PMID: 34278583 PMCID: PMC8763953 DOI: 10.1002/jcp.30528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/01/2021] [Accepted: 07/06/2021] [Indexed: 01/03/2023]
Abstract
Diacylglycerol kinase (DGK), a lipid kinase, catalyzes the conversion of diacylglycerol (DAG) to phosphatidic acid, thereby terminating DAG-mediated signaling by Gq-coupled receptors that regulate contraction of airway smooth muscle (ASM). A previous study from our laboratory demonstrated that DGK inhibition or genetic ablation leads to reduced ASM contraction and provides protection for allergen-induced airway hyperresponsiveness. However, the mechanism by which DGK regulates contractile signaling in ASM is not well established. Herein, we investigated the role of prorelaxant cAMP-protein kinase A (PKA) signaling in DGK-mediated regulation of ASM contraction. Pretreatment of human ASM cells with DGK inhibitor I activated PKA as demonstrated by the phosphorylation of PKA substrates, VASP, Hsp20, and CREB, which was abrogated when PKA was inhibited pharmacologically or molecularly using overexpression of the PKA inhibitor peptide, PKI. Furthermore, inhibition of DGK resulted in induction of cyclooxygenase (COX) and generation of prostaglandin E2 (PGE2 ) with concomitant activation of Gs-cAMP-PKA signaling in ASM cells in an autocrine/paracrine fashion. Inhibition of protein kinase C (PKC) or extracellular-signal-regulated kinase (ERK) attenuated DGK-mediated production of PGE2 and activation of cAMP-PKA signaling in human ASM cells, suggesting that inhibition of DGK activates the COX-PGE2 pathway in a PKC-ERK-dependent manner. Finally, DGK inhibition-mediated attenuation of contractile agonist-induced phosphorylation of myosin light chain 20 (MLC-20), a marker of ASM contraction, involves COX-mediated cAMP production and PKA activation in ASM cells. Collectively these findings establish a novel mechanism by which DGK regulates ASM contraction and further advances DGK as a potential therapeutic target to provide effective bronchoprotection in asthma.
Collapse
Affiliation(s)
- Santosh K. Yadav
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA 19107
| | - Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA 19107
| | - Sushrut D. Shah
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA 19107
| | - Reynold A. Panettieri
- Rutgers Institute for Translational Medicine & Science, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901
| | - Taku Kambayashi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Raymond B. Penn
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA 19107
| | - Deepak A. Deshpande
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA 19107.,Corresponding author Deepak Deshpande, PhD, Professor, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, USA 19107,
| |
Collapse
|
5
|
Sharma P, Penn RB. Can GPCRs Be Targeted to Control Inflammation in Asthma? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:1-20. [PMID: 34019260 DOI: 10.1007/978-3-030-68748-9_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Historically, the drugs used to manage obstructive lung diseases (OLDs), asthma, and chronic obstructive pulmonary disease (COPD) either (1) directly regulate airway contraction by blocking or relaxing airway smooth muscle (ASM) contraction or (2) indirectly regulate ASM contraction by inhibiting the principal cause of ASM contraction/bronchoconstriction and airway inflammation. To date, these tasks have been respectively assigned to two diverse drug types: agonists/antagonists of G protein-coupled receptors (GPCRs) and inhaled or systemic steroids. These two types of drugs "stay in their lane" with respect to their actions and consequently require the addition of the other drug to effectively manage both inflammation and bronchoconstriction in OLDs. Indeed, it has been speculated that safety issues historically associated with beta-agonist use (beta-agonists activate the beta-2-adrenoceptor (β2AR) on airway smooth muscle (ASM) to provide bronchoprotection/bronchorelaxation) are a function of pro-inflammatory actions of β2AR agonism. Recently, however, previously unappreciated roles of various GPCRs on ASM contractility and on airway inflammation have been elucidated, raising the possibility that novel GPCR ligands targeting these GPCRs can be developed as anti-inflammatory therapeutics. Moreover, we now know that many GPCRs can be "tuned" and not just turned "off" or "on" to specifically activate the beneficial therapeutic signaling a receptor can transduce while avoiding detrimental signaling. Thus, the fledging field of biased agonism pharmacology has the potential to turn the β2AR into an anti-inflammatory facilitator in asthma, possibly reducing or eliminating the need for steroids.
Collapse
Affiliation(s)
- Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy, & Critical Care Medicine Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College Thomas Jefferson University, Philadelphia, PA, USA
| | - Raymond B Penn
- Center for Translational Medicine, Division of Pulmonary, Allergy, & Critical Care Medicine Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
6
|
Penn RB. Mast cells in asthma: Here I am, stuck in the middle with you. Eur Respir J 2020; 56:56/1/2001337. [PMID: 32616549 PMCID: PMC7643049 DOI: 10.1183/13993003.01337-2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022]
|
7
|
Michael JV, Gavrila A, Nayak AP, Pera T, Liberato JR, Polischak SR, Shah SD, Deshpande DA, Penn RB. Cooperativity of E-prostanoid receptor subtypes in regulating signaling and growth inhibition in human airway smooth muscle. FASEB J 2019; 33:4780-4789. [PMID: 30601680 DOI: 10.1096/fj.201801959r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Prostaglandin E2 (PGE2) is produced in the airway during allergic lung inflammation and both promotes and inhibits features of asthma pathology. These mixed effects relate to 4 E-prostanoid (EP) receptor subtypes (EP1, 2, 3 and 4) expressed at different levels on different resident and infiltrating airway cells. Although studies have asserted both EP2 and EP4 expression in human airway smooth muscle (HASM), a recent study asserted EP4 to be the functionally dominant EP subtype in HASM. Herein, we employ recently-developed subtype-selective ligands to investigate singular or combined EP2 and EP4 receptor activation in regulating HASM signaling and proliferation. The subtype specificity of ONO-AE1-259-01 (EP2 agonist) and ONO-AE1-329 (EP4 agonist) was first demonstrated in human embryonic kidney 293 cells stably expressing different EP receptor subtypes. EP receptor knockdown and subtype-selective antagonists demonstrated EP2 and EP4 receptor responsiveness in HASM cells to the specific ONO compounds, whereas PGE2 appeared to preferentially signal via the EP4 receptor. Both singular EP2 and EP4 receptor agonists inhibited HASM proliferation, and combined EP2 and EP4 receptor agonism exhibited positive cooperativity in both chronic Gs-mediated signaling and inhibiting HASM proliferation. These findings suggest both EP2 and EP4 are functionally important in HASM, and their combined targeting optimally inhibits airway smooth muscle proliferation.-Michael, J. V. Gavrila, A., Nayak, A. P., Pera, T., Liberato, J. R., Polischak, S. R., Shah, S. D., Deshpande, D. A., Penn, R. B. Cooperativity of E-prostanoid receptor subtypes in regulating signaling and growth inhibition in human airway smooth muscle.
Collapse
Affiliation(s)
- James V Michael
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adelina Gavrila
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ajay P Nayak
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Tonio Pera
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jennifer R Liberato
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Steven R Polischak
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sushrut D Shah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Deepak A Deshpande
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Raymond B Penn
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Center for Translational Medicine, Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
8
|
Nayak AP, Deshpande DA, Penn RB. New targets for resolution of airway remodeling in obstructive lung diseases. F1000Res 2018; 7. [PMID: 29904584 PMCID: PMC5981194 DOI: 10.12688/f1000research.14581.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2018] [Indexed: 12/17/2022] Open
Abstract
Airway remodeling (AR) is a progressive pathological feature of the obstructive lung diseases, including asthma and chronic obstructive pulmonary disease (COPD). The pathology manifests itself in the form of significant, progressive, and (to date) seemingly irreversible changes to distinct respiratory structural compartments. Consequently, AR correlates with disease severity and the gradual decline in pulmonary function associated with asthma and COPD. Although current asthma/COPD drugs manage airway contraction and inflammation, none of these effectively prevent or reverse features of AR. In this review, we provide a brief overview of the features and putative mechanisms affecting AR. We further discuss recently proposed strategies with promise for deterring or treating AR.
Collapse
Affiliation(s)
- Ajay P Nayak
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, USA
| | - Deepak A Deshpande
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, USA
| | - Raymond B Penn
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
9
|
Kouatly O, Eleftheriou P, Petrou A, Hadjipavlou-Litina D, Geronikaki A. Docking assisted design of novel 4-adamantanyl-2-thiazolylimino-5-arylidene-4-thiazolidinones as potent NSAIDs. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2018; 29:83-101. [PMID: 29299942 DOI: 10.1080/1062936x.2017.1410220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 11/23/2017] [Indexed: 06/07/2023]
Abstract
Docking analysis was used to predict the effectiveness of adamantanyl insertion in improving cycloxygenase/lipoxygenase (COX/LOX) inhibitory action of previously tested 2-thiazolylimino-5-arylidene-4-thiazolidinones. The crystal structure data of human 5-LOX (3O8Y), ovine COX-1 (1EQH) and mouse COX-2 (3ln1) were used for docking analysis. All docking calculations were carried out using AutoDock 4.2 software. Following prediction results, 11 adamantanyl derivatives were synthesized and evaluated for biological action. Prediction evaluations correlated well with experimental biological results. Comparison of the novel adamantanyl derivatives with the 2-thiazolylimino-5-arylidene-4-thiazolidinones previously tested showed that insertion of the adamantanyl group led to the production of more potent COX-1 inhibitors, as well as LOX inhibitors (increased activity from 200% to 560%). Five compounds out of the 11 exhibited better activity than naproxen; while nine out of 11 showed better activity than NDGA and seven compounds possessed better anti-inflammatory activity than indomethacin.
Collapse
Affiliation(s)
- O Kouatly
- a Department of Pharmaceutical Chemistry, School of Pharmacy , Aristotle University of Thessaloniki , Greece
| | - Ph Eleftheriou
- b Department of Medical Laboratories, School of Health and Care Professions , Alexandrion Technological Educational Institute of Thessaloniki , Greece
| | - A Petrou
- a Department of Pharmaceutical Chemistry, School of Pharmacy , Aristotle University of Thessaloniki , Greece
| | - D Hadjipavlou-Litina
- a Department of Pharmaceutical Chemistry, School of Pharmacy , Aristotle University of Thessaloniki , Greece
| | - A Geronikaki
- a Department of Pharmaceutical Chemistry, School of Pharmacy , Aristotle University of Thessaloniki , Greece
| |
Collapse
|
10
|
Mohamed NA, Davies RP, Lickiss PD, Ahmetaj-Shala B, Reed DM, Gashaw HH, Saleem H, Freeman GR, George PM, Wort SJ, Morales-Cano D, Barreira B, Tetley TD, Chester AH, Yacoub MH, Kirkby NS, Moreno L, Mitchell JA. Chemical and biological assessment of metal organic frameworks (MOFs) in pulmonary cells and in an acute in vivo model: relevance to pulmonary arterial hypertension therapy. Pulm Circ 2017; 7:643-653. [PMID: 28447910 PMCID: PMC5841901 DOI: 10.1177/2045893217710224] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and debilitating condition. Despite promoting vasodilation, current drugs have a therapeutic window within which they are limited by systemic side effects. Nanomedicine uses nanoparticles to improve drug delivery and/or reduce side effects. We hypothesize that this approach could be used to deliver PAH drugs avoiding the systemic circulation. Here we report the use of iron metal organic framework (MOF) MIL-89 and PEGylated MIL-89 (MIL-89 PEG) as suitable carriers for PAH drugs. We assessed their effects on viability and inflammatory responses in a wide range of lung cells including endothelial cells grown from blood of donors with/without PAH. Both MOFs conformed to the predicted structures with MIL-89 PEG being more stable at room temperature. At concentrations up to 10 or 30 µg/mL, toxicity was only seen in pulmonary artery smooth muscle cells where both MOFs reduced cell viability and CXCL8 release. In endothelial cells from both control donors and PAH patients, both preparations inhibited the release of CXCL8 and endothelin-1 and in macrophages inhibited inducible nitric oxide synthase activity. Finally, MIL-89 was well-tolerated and accumulated in the rat lungs when given in vivo. Thus, the prototypes MIL-89 and MIL-89 PEG with core capacity suitable to accommodate PAH drugs are relatively non-toxic and may have the added advantage of being anti-inflammatory and reducing the release of endothelin-1. These data are consistent with the idea that these materials may not only be useful as drug carriers in PAH but also offer some therapeutic benefit in their own right.
Collapse
Affiliation(s)
- Nura A Mohamed
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK.,2 Heart Science Centre at Harefield Hospital, Harefield, UK.,3 Qatar Foundation Research and Development Division, Doha, Qatar
| | - Robert P Davies
- 4 Department of Chemistry, South Kensington Campus, Imperial College, London, UK
| | - Paul D Lickiss
- 4 Department of Chemistry, South Kensington Campus, Imperial College, London, UK
| | - Blerina Ahmetaj-Shala
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Daniel M Reed
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Hime H Gashaw
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Hira Saleem
- 4 Department of Chemistry, South Kensington Campus, Imperial College, London, UK
| | - Gemma R Freeman
- 4 Department of Chemistry, South Kensington Campus, Imperial College, London, UK
| | - Peter M George
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Stephen J Wort
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Daniel Morales-Cano
- 5 Department of Pharmacology, Faculty of Medicine, Universidad Complutense de Madrid- Instituto de Investigacion Sanitaria Gregorio Marañón (IiSGM), Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Bianca Barreira
- 5 Department of Pharmacology, Faculty of Medicine, Universidad Complutense de Madrid- Instituto de Investigacion Sanitaria Gregorio Marañón (IiSGM), Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Teresa D Tetley
- 6 Lung Cell Biology Group, National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Magdi H Yacoub
- 2 Heart Science Centre at Harefield Hospital, Harefield, UK
| | - Nicholas S Kirkby
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | - Laura Moreno
- 5 Department of Pharmacology, Faculty of Medicine, Universidad Complutense de Madrid- Instituto de Investigacion Sanitaria Gregorio Marañón (IiSGM), Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Jane A Mitchell
- 1 Department of Cardiothoracic Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
11
|
Rumzhum NN, Ammit AJ. Cyclooxygenase 2: its regulation, role and impact in airway inflammation. Clin Exp Allergy 2016; 46:397-410. [PMID: 26685098 DOI: 10.1111/cea.12697] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cyclooxygenase 2 (COX-2: official gene symbol - PTGS2) has long been regarded as playing a pivotal role in the pathogenesis of airway inflammation in respiratory diseases including asthma. COX-2 can be rapidly and robustly expressed in response to a diverse range of pro-inflammatory cytokines and mediators. Thus, increased levels of COX-2 protein and prostanoid metabolites serve as key contributors to pathobiology in respiratory diseases typified by dysregulated inflammation. But COX-2 products may not be all bad: prostanoids can exert anti-inflammatory/bronchoprotective functions in airways in addition to their pro-inflammatory actions. Herein, we outline COX-2 regulation and review the diverse stimuli known to induce COX-2 in the context of airway inflammation. We discuss some of the positive and negative effects that COX-2/prostanoids can exert in in vitro and in vivo models of airway inflammation, and suggest that inhibiting COX-2 expression to repress airway inflammation may be too blunt an approach; because although it might reduce the unwanted effects of COX-2 activation, it may also negate the positive effects. Evidence suggests that prostanoids produced via COX-2 upregulation show diverse actions (and herein we focus on prostaglandin E2 as a key example); these can be either beneficial or deleterious and their impact on respiratory disease can be dictated by local concentration and specific interaction with individual receptors. We propose that understanding the regulation of COX-2 expression and associated receptor-mediated functional outcomes may reveal number of critical steps amenable to pharmacological intervention. These may prove invaluable in our quest towards future development of novel anti-inflammatory pharmacotherapeutic strategies for the treatment of airway diseases.
Collapse
Affiliation(s)
- N N Rumzhum
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| | - A J Ammit
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
12
|
Hameed AD, Ovais S, Yaseen R, Rathore P, Samim M, Singh S, Sharma K, Akhtar M, Javed K. Synthesis and Biological Evaluation of New Phthalazinone Derivatives as Anti-Inflammatory and Anti-Proliferative Agents. Arch Pharm (Weinheim) 2016; 349:150-9. [DOI: 10.1002/ardp.201500336] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/01/2015] [Accepted: 12/04/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Alhamzah Dh. Hameed
- Department of Chemistry, Faculty of Science; Jamia Hamdard (Hamdard University); New Delhi India
| | - Syed Ovais
- Department of Chemistry, Faculty of Science; Jamia Hamdard (Hamdard University); New Delhi India
| | - Raed Yaseen
- Department of Chemistry, Faculty of Science; Jamia Hamdard (Hamdard University); New Delhi India
| | - Pooja Rathore
- Department of Chemistry, Faculty of Science; Jamia Hamdard (Hamdard University); New Delhi India
| | - Mohammed Samim
- Department of Chemistry, Faculty of Science; Jamia Hamdard (Hamdard University); New Delhi India
| | - Surender Singh
- Department of Pharmacology; All India Institute of Medical Science; New Delhi India
| | - Kalicharan Sharma
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Drug Design and Medicinal Chemistry Laboratory; Jamia Hamdard (Hamdard University); New Delhi India
| | - Mymona Akhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Drug Design and Medicinal Chemistry Laboratory; Jamia Hamdard (Hamdard University); New Delhi India
| | - Kalim Javed
- Department of Chemistry, Faculty of Science; Jamia Hamdard (Hamdard University); New Delhi India
| |
Collapse
|
13
|
Prabhala P, Bunge K, Rahman MM, Ge Q, Clark AR, Ammit AJ. Temporal regulation of cytokine mRNA expression by tristetraprolin: dynamic control by p38 MAPK and MKP-1. Am J Physiol Lung Cell Mol Physiol 2015; 308:L973-80. [PMID: 25724669 DOI: 10.1152/ajplung.00219.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 02/22/2015] [Indexed: 01/16/2023] Open
Abstract
Cytokines drive many inflammatory diseases, including asthma. Understanding the molecular mechanisms responsible for cytokine secretion will allow us to develop novel strategies to repress inflammation in the future. Harnessing the power of endogenous anti-inflammatory proteins is one such strategy. In this study, we investigate the p38 MAPK-mediated regulatory interaction of two anti-inflammatory proteins, mitogen-activated protein kinase phosphatase 1 (MKP-1) and tristetraprolin (TTP), in the context of asthmatic inflammation. Using primary cultures of airway smooth muscle cells in vitro, we explored the temporal regulation of IL-6 cytokine mRNA expression upon stimulation with TNF-α. Intriguingly, the temporal profile of mRNA expression was biphasic. This was not due to COX-2-derived prostanoid upregulation, increased expression of NLRP3 inflammasome components, or upregulation of the cognate receptor for TNF-α-TNFR1. Rather, the biphasic nature of TNF-α-induced IL-6 mRNA expression was regulated temporally by the RNA-destabilizing molecule, TTP. Importantly, TTP function is controlled by p38 MAPK, and our study reveals that its expression in airway smooth muscle cells is p38 MAPK-dependent and its anti-inflammatory activity is also controlled by p38 MAPK-mediated phosphorylation. MKP-1 is a MAPK deactivator; thus, by controlling p38 MAPK phosphorylation status in a temporally distinct manner, MKP-1 ensures that TTP is expressed and made functional at precisely the correct time to repress cytokine expression. Together, p38 MAPK, MKP-1, and TTP may form a regulatory network that exerts significant control on cytokine secretion in proasthmatic inflammation through precise temporal signaling.
Collapse
Affiliation(s)
- Pavan Prabhala
- Faculty of Pharmacy, University of Sydney, New South Wales, Australia
| | - Kristin Bunge
- Faculty of Pharmacy, University of Sydney, New South Wales, Australia
| | | | - Qi Ge
- Woolcock Institute of Medical Research, University of Sydney, New South Wales, Australia; and
| | - Andrew R Clark
- Centre for Translational Inflammation Research, School of Immunity and Infection, University of Birmingham, Edgbaston, United Kingdom
| | - Alaina J Ammit
- Faculty of Pharmacy, University of Sydney, New South Wales, Australia;
| |
Collapse
|
14
|
Thromboxane A2 exerts promoting effects on cell proliferation through mediating cyclooxygenase-2 signal in lung adenocarcinoma cells. J Cancer Res Clin Oncol 2014; 140:375-86. [DOI: 10.1007/s00432-013-1573-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 12/16/2013] [Indexed: 12/15/2022]
|
15
|
Billington CK, Ojo OO, Penn RB, Ito S. cAMP regulation of airway smooth muscle function. Pulm Pharmacol Ther 2013; 26:112-20. [PMID: 22634112 PMCID: PMC3574867 DOI: 10.1016/j.pupt.2012.05.007] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/14/2012] [Accepted: 05/17/2012] [Indexed: 12/11/2022]
Abstract
Agonists activating β(2)-adrenoceptors (β(2)ARs) on airway smooth muscle (ASM) are the drug of choice for rescue from acute bronchoconstriction in patients with both asthma and chronic obstructive pulmonary disease (COPD). Moreover, the use of long-acting β-agonists combined with inhaled corticosteroids constitutes an important maintenance therapy for these diseases. β-Agonists are effective bronchodilators due primarily to their ability to antagonize ASM contraction. The presumed cellular mechanism of action involves the generation of intracellular cAMP, which in turn can activate the effector molecules cAMP-dependent protein kinase (PKA) and Epac. Other agents such as prostaglandin E(2) and phosphodiesterase inhibitors that also increase intracellular cAMP levels in ASM, can also antagonize ASM contraction, and inhibit other ASM functions including proliferation and migration. Therefore, β(2)ARs and cAMP are key players in combating the pathophysiology of airway narrowing and remodeling. However, limitations of β-agonist therapy due to drug tachyphylaxis related to β(2)AR desensitization, and recent findings regarding the manner in which β(2)ARs and cAMP signal, have raised new and interesting questions about these well-studied molecules. In this review we discuss current concepts regarding β(2)ARs and cAMP in the regulation of ASM cell functions and their therapeutic roles in asthma and COPD.
Collapse
Affiliation(s)
- Charlotte K Billington
- Division of Therapeutics and Molecular Medicine, The University of Nottingham, Nottingham NG7 2UH, UK.
| | | | | | | |
Collapse
|
16
|
Brivio I, Buccellati C, Fumagalli F, Hodge J, Casagrande C, Folco GC, Sala A. The pulmonary pharmacology of [4-methoxy-N1-(4-trans-nitrooxycyclohexyl)-N3-(3-pyridinylmethyl)-1,3-benzenedicarboxamide] (2NTX-99), an anti-atherotrombotic compound with therapeutic potential in pathological conditions that target lung vasculature. Prostaglandins Other Lipid Mediat 2012; 98:116-21. [PMID: 22342851 DOI: 10.1016/j.prostaglandins.2012.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 01/23/2012] [Accepted: 01/26/2012] [Indexed: 10/14/2022]
Abstract
The pharmacological activity of 2NTX-99 ([4-methoxy-N1-(4-trans-nitrooxycyclohexyl)-N3-(3-pyridinylmethyl)-1,3-benzenedicarboxamide]) was investigated in vitro in the intact, rat pulmonary vasculature and in guinea pig airways. Rat lungs were perfused at constant flow and changes in vascular tone recorded. Challenge with the TXA₂ analogue 9,11-dideoxy-9α11α-methanoepoxy ProstaglandinF₂ (U46619, 0.5 μM) increased vessel tone (32.48±1.5 vs 13.13±0.56 mmHg; n=12). 2NTX-99 (0.1-100 μM; n=5), caused a concentration-dependent relaxation, prevented by 1H-[1,2,4]oxadiazolo[4,3,-a]quinoxalin-1-one (ODQ, 10 μM, n=4), an inhibitor of soluble guanylate cyclase. Acetylcholine (0.1-10 μM; n=3) and a reference NO-donor, isosorbide-5-mononitrate (5-100 μM; n=4), were ineffective. Intraluminal perfusion of washed human platelets (2 × 10⁸ cells/ml) increased intravascular pressure after challenge with arachidonic acid (AA, 2 μM; n=5), an increase abolished by acetylsalicylic acid and significantly reduced by 2NTX-99 (40 μM; n=5). TXB₂ in the lung perfusate was detected after platelet activation, 2NTX-99 inhibited TXA₂ synthesis (6.45±0.6 and 1.10±0.2 ng/ml, respectively). 2NTX-99 did not alter central or peripheral airway responsiveness to Histamine (0.001-300 μM; n=6), U46619 (0.001-3 μM, n=3) or LTD₄ (1 pM-1 μM; n=6). 2NTX-99 vasodilates the pulmonary vasculature via the release of nitric oxide (NO) and reduces intraluminal, AA-induced, TXA₂ formation. The combined activity of 2NTX-99 as an NO-donor and a TXA₂-synthesis inhibitor provides strong support for its potential therapeutic use in pathologies of the pulmonary vascular bed (e.g. pulmonary hypertension).
Collapse
Affiliation(s)
- I Brivio
- Department of Pharmacological Sciences, School of Pharmacy, University of Milan, Via Balzaretti 9, 20133 Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
17
|
Peroxisome proliferator activated receptor ligands as regulators of airway inflammation and remodelling in chronic lung disease. PPAR Res 2011; 2007:14983. [PMID: 18000530 PMCID: PMC2065911 DOI: 10.1155/2007/14983] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Accepted: 06/11/2007] [Indexed: 11/20/2022] Open
Abstract
Inflammation is a major component in the pathology of chronic lung diseases, including asthma. Anti-inflammatory treatment with corticosteroids is not effective in all patients. Thus, new therapeutic options are required to control diverse cellular functions that are currently not optimally targeted by these drugs in order to inhibit inflammation and its sequelae in lung disease. Peroxisome proliferator activated receptors (PPARs), originally characterised as regulators of lipid and glucose metabolism, offer marked potential in this respect. PPARs are expressed in both lung infiltrating and resident immune and inflammatory cells, as well as in resident and structural cells in the lungs, and play critical roles in the regulation of airway inflammation. In vitro, endogenous and synthetic ligands for PPARs regulate expression and release of proinflammatory cytokines and chemoattractants, and cell proliferation and survival. In murine models of allergen-induced inflammation, PPARα and PPARγ ligands reduce the influx of inflammatory cells, cytokine and mucus production, collagen deposition, and airways hyperresponsiveness. The activity profiles of PPAR ligands differ to corticosteroids, supporting the hypothesis that PPARs comprise additional therapeutic targets to mimimise the contribution of inflammation to airway remodelling and dysfunction.
Collapse
|
18
|
Harding P, LaPointe MC. Prostaglandin E2 increases cardiac fibroblast proliferation and increases cyclin D expression via EP1 receptor. Prostaglandins Leukot Essent Fatty Acids 2011; 84:147-52. [PMID: 21342756 PMCID: PMC3071899 DOI: 10.1016/j.plefa.2011.01.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 01/12/2011] [Accepted: 01/12/2011] [Indexed: 10/18/2022]
Abstract
PGE(2) affects growth of many cell types. Thus, we hypothesized that PGE(2) would stimulate growth of cardiac fibroblasts. To test our hypothesis we used neonatal rat ventricular fibroblasts (NVF). RT-PCR demonstrated the presence of all 4 PGE(2) receptor (EPs) mRNAs in NVF. Using flow cytometry, we found that PGE(2) decreased the percentage of cells in G0/G1 and increased the number of cells in S phase. PGE(2) also increased expression of cyclin D3, a known regulator of the cell cycle and this effect was mimicked by the EP1/EP3 agonist sulprostone. Next, we found that treatment of NVF with PGE(2) increased phosphorylation of p42/44 MAPK and Akt and that PGE(2)-stimulation of cyclin D3 was antagonized with both a MEK inhibitor and a PI3 kinase inhibitor. In conclusion, PGE(2) stimulates cardiac fibroblast proliferation via EP1 and/or EP3, p42/44 MAPK and Akt-regulation of cyclin D3. These results may be relevant to cardiac fibrosis.
Collapse
Affiliation(s)
- Pamela Harding
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI 48202, USA.
| | | |
Collapse
|
19
|
Mori A, Ito S, Morioka M, Aso H, Kondo M, Sokabe M, Hasegawa Y. Effects of specific prostanoid EP receptor agonists on cell proliferation and intracellular Ca(2+) concentrations in human airway smooth muscle cells. Eur J Pharmacol 2011; 659:72-8. [PMID: 21397595 DOI: 10.1016/j.ejphar.2011.03.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Revised: 02/17/2011] [Accepted: 03/01/2011] [Indexed: 10/18/2022]
Abstract
Increased airway smooth muscle mass due to cell proliferation contributes to airway hyper-responsiveness and remodeling in patients with asthma. Prostaglandin E2 (PGE2) inhibits proliferation of airway smooth muscle cells, but the role of prostanoid EP receptor subtypes in mechanisms involved has not been fully elucidated yet. We investigated the effects of specific prostanoid EP receptor agonists on cell proliferation and intracellular Ca(2+) concentrations ([Ca(2+)]i) in human airway smooth muscle cells. Cell numbers were assessed by mitochondria-dependent reduction of 4-[3-(4-lodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1, 3-benzene disulfonate to formazan (WST-1 assay). RT-PCR data showed that human airway smooth muscle cells express EP2, EP3, and EP4 but not EP1 receptor mRNA. PGE2 (1nM-1μM) inhibited cell proliferation induced by 5% fetal bovine serum (FBS) in a concentration-dependent manner. (16S)-9-deoxy-9β-chloro-15-deoxy-16-hydroxy-17, 17-trimethylene-19, 20-didehydro PGE2 sodium salt (ONO-AE1-259-01; EP2 receptor agonist) and 16-(3-methoxymethyl)phenyl-ω-tetranor-3,7-dithia PGE2 (ONO-AE1-329; EP4 receptor agonist) inhibited the 5% FBS-induced cell proliferation. ONO-AE1-259-01 and ONO-AE1-329 also significantly increased the cytosolic cAMP levels. In contrast, 11,15-O-dimethyl PGE2 (ONO-AE-248; EP3 receptor agonist) elicited an oscillatory increase in [Ca(2+)]i but did not affect the cell growth or cAMP levels. [(17S)-2,5-ethano-6-oxo-17,20-dimethyl PGE1] (ONO-DI-004; EP1 receptor agonist) did not affect cell growth, cAMP levels, or [Ca(2+)]i. In conclusion, PGE2 inhibits FBS-induced cell proliferation mostly via EP2 and EP4 receptor activation and subsequent cAMP elevation. The EP3 receptor agonist causes an increase in [Ca(2+)]i without affecting cell growth. There is no functional expression of the EP1 receptor. Research on prostanoid EP receptors may lead to novel therapeutic strategies for treatment of asthma.
Collapse
Affiliation(s)
- Akemi Mori
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Satoru Ito
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masataka Morioka
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hiromichi Aso
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masashi Kondo
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Masahiro Sokabe
- Department of Physiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshinori Hasegawa
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
20
|
Balakumar C, Lamba P, Kishore DP, Narayana BL, Rao KV, Rajwinder K, Rao AR, Shireesha B, Narsaiah B. Synthesis, anti-inflammatory evaluation and docking studies of some new fluorinated fused quinazolines. Eur J Med Chem 2010; 45:4904-13. [PMID: 20800934 DOI: 10.1016/j.ejmech.2010.07.063] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 07/22/2010] [Accepted: 07/29/2010] [Indexed: 11/28/2022]
Abstract
A series of novel 8/10-trifluoromethyl-substituted-imidazo[1,2-c] quinazolines have been synthesized and evaluated in vivo (rat paw edema) for their anti-inflammatory activity and in silico (docking studies) to recognize the hypothetical binding motif of the title compounds with the cyclooxygenase isoenzymes (COX-1 and COX-2) employing GOLD (CCDC, 4.0.1 version) software. The compounds, 9b and 10b, were found to have good anti-inflammatory activity [around 80% of the standard: indomethacin]. The binding mode of the title compounds has been proposed based on the docking studies.
Collapse
Affiliation(s)
- C Balakumar
- Centre with Potential for Excellence in Biomedical Sciences, Panjab University, Chandigarh 160 014, India
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Moore BB, Peters-Golden M. Opposing roles of leukotrienes and prostaglandins in fibrotic lung disease. Expert Rev Clin Immunol 2010; 2:87-100. [PMID: 20477090 DOI: 10.1586/1744666x.2.1.87] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Lung fibrosis is a devastating disease that involves a variable degree of inflammation, alveolar epithelial injury, fibroblast hyperplasia and the deposition of extracellular matrix. Standard therapies that consist of corticosteroids and immunosuppressive agents offer little benefit and most patients experience a progressive deterioration in lung function which is ultimately fatal within 2-5 years of diagnosis. New pathogenetic insights and therapeutic approaches are badly needed. Eicosanoids are lipid mediators derived from arachidonic acid metabolism, the best studied of which are prostaglandins and leukotrienes. Although these mediators are primarily known for their roles in asthma, pain, fever and vascular responses, they also exert relevant effects on immune and inflammatory cells as well as structural cells such as epithelial cells and fibroblasts - cell types which participate in fibrogenesis. In general, leukotrienes promote while prostaglandin E(2) opposes fibrogenic responses. Lung fibrosis is associated with increased production of leukotrienes and decreased production of prostaglandin E(2). Furthermore, responses to prostaglandin E(2) are altered in fibrotic conditions. This review highlights the role of this leukotriene/prostaglandin imbalance in the evolution of fibrotic lung disease, offers insights into the mechanisms that underlie the dysregulated responses and discusses approaches for therapeutic targeting of eicosanoids in these conditions.
Collapse
Affiliation(s)
- Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, 6220 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI 48109-0642, USA.
| | | |
Collapse
|
22
|
Donnellan PD, Kinsella BT. Immature and mature species of the human Prostacyclin Receptor are ubiquitinated and targeted to the 26S proteasomal or lysosomal degradation pathways, respectively. J Mol Signal 2009; 4:7. [PMID: 19781057 PMCID: PMC2760523 DOI: 10.1186/1750-2187-4-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Accepted: 09/25/2009] [Indexed: 12/22/2022] Open
Abstract
Background The human prostacyclin receptor (hIP) undergoes agonist-induced phosphorylation, desensitisation and internalisation and may be recycled to the plasma membrane or targeted for degradation by, as yet, unknown mechanism(s). Results Herein it was sought to investigate the turnover of the hIP under basal conditions and in response to cicaprost stimulation. It was established that the hIP is subject to low-level basal degradation but, following agonist stimulation, degradation is substantially enhanced. Inhibition of the lysosomal pathway prevented basal and agonist-induced degradation of the mature species of the hIP (46-66 kDa). Conversely, inhibition of the proteasomal pathway had no effect on levels of the mature hIP but led to time-dependent accumulation of four newly synthesised immature species (38-44 kDa). It was established that both the mature and immature species of the hIP may be polyubiquitinated and this modification may be required for lysosomal sorting of the mature, internalised receptors and for degradation of the immature receptors by the 26S proteasomes through the ER-associated degradation (ERAD) process, respectively. Moreover, these data substantially advance knowledge of the factors regulating processing and maturation of the hIP, a complex receptor subject to multiple post-translational modifications including N-glycosylation, phosphorylation, isoprenylation, palmitoylation, in addition to polyubiquitination, as determined herein. Conclusion These findings indicate that the hIP is post-translationally modified by ubiquitination, which targets the immature species to the 26S proteasomal degradation pathway and the mature species to the lysosomal degradation pathway.
Collapse
Affiliation(s)
- Peter D Donnellan
- School of Biomolecular and Biomedical Sciences, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | | |
Collapse
|
23
|
Tliba O, Panettieri RA. Noncontractile functions of airway smooth muscle cells in asthma. Annu Rev Physiol 2009; 71:509-35. [PMID: 18851708 DOI: 10.1146/annurev.physiol.010908.163227] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although pivotal in regulating bronchomotor tone in asthma, airway smooth muscle (ASM) also modulates airway inflammation and undergoes hypertrophy and hyperplasia, contributing to airway remodeling in asthma. ASM myocytes secrete or express a wide array of immunomodulatory mediators in response to extracellular stimuli, and in chronic severe asthma, increases in ASM mass may render the airway irreversibly obstructed. Although the mechanisms by which ASM secretes cytokines and chemokines are the same as those regulating immune cells, there exist unique ASM signaling pathways that may provide novel therapeutic targets. This review provides an overview of our current understanding of the proliferative as well as the synthetic properties of ASM.
Collapse
Affiliation(s)
- Omar Tliba
- Pulmonary, Allergy and Critical Care Division, Airways Biology Initiative, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
24
|
Clarke DL, Dakshinamurti S, Larsson AK, Ward JE, Yamasaki A. Lipid metabolites as regulators of airway smooth muscle function. Pulm Pharmacol Ther 2008; 22:426-35. [PMID: 19114116 DOI: 10.1016/j.pupt.2008.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 11/18/2008] [Accepted: 12/10/2008] [Indexed: 02/02/2023]
Abstract
Compelling evidence identifies airway smooth muscle (ASM) not only as a target but also a cellular source for a diverse range of mediators underlying the processes of airway narrowing and airway hyperresponsiveness in diseases such as asthma. These include the growing family of plasma membrane phospholipid-derived polyunsaturated fatty acids broadly characterised by the prostaglandins, leukotrienes, lipoxins, isoprostanes and lysophospholipids. In this review, we describe the enzymatic and non-enzymatic biosynthetic pathways of these lipid mediators and how these are influenced by drug treatment, oxidative stress and airways disease. Additionally, we outline their cognate receptors, many of which are expressed by ASM. We describe potential deleterious and protective roles for these lipid mediators in airway inflammatory and remodelling processes by describing their effects on diverse functions of ASM in asthma that have the potential to contribute to asthma pathogenesis and symptoms. These functions include contractile tone development, cytokine and extracellular matrix production, and cellular proliferation and migration.
Collapse
Affiliation(s)
- Deborah L Clarke
- Respiratory Pharmacology, National Heart and Lung Institute, Imperial College, London, UK
| | | | | | | | | |
Collapse
|
25
|
Kaur M, Holden NS, Wilson SM, Sukkar MB, Chung KF, Barnes PJ, Newton R, Giembycz MA. Effect of beta2-adrenoceptor agonists and other cAMP-elevating agents on inflammatory gene expression in human ASM cells: a role for protein kinase A. Am J Physiol Lung Cell Mol Physiol 2008; 295:L505-14. [PMID: 18586957 DOI: 10.1152/ajplung.00046.2008] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In diseases such as asthma, airway smooth muscle (ASM) cells play a synthetic role by secreting inflammatory mediators such as granulocyte-macrophage colony-stimulating factor (GM-CSF), IL-6, or IL-8 and by expressing surface adhesion molecules, including ICAM-1. In the present study, PGE(2), forskolin, and short-acting (salbutamol) and long-acting (salmeterol and formoterol) beta(2)-adrenoceptor agonists reduced the expression of ICAM-1 and the release of GM-CSF evoked by IL-1beta in ASM cells. IL-1beta-induced IL-8 release was also repressed by PGE(2) and forskolin, whereas the beta(2)-adrenoceptor agonists were ineffective. In each case, repression of these inflammatory indexes was prevented by adenoviral overexpression of PKIalpha, a highly selective PKA inhibitor. These data indicate a PKA-dependent mechanism of repression and suggest that agents that elevate intracellular cAMP, and thereby activate PKA, may have a widespread anti-inflammatory effect in ASM cells. Since ICAM-1 and GM-CSF are highly NF-kappaB-dependent genes, we used an adenoviral-delivered NF-kappaB-dependent luciferase reporter to examine the effects of forskolin and the beta(2)-adrenoceptor agonists on NF-kappaB activation. There was no effect on luciferase activity measured in the presence of forskolin or beta(2)-adrenoceptor agonists. This finding is consistent with the observation that IL-1beta-induced expression of IL-6, a known NF-kappaB-dependent gene in ASM, was also unaffected by beta(2)-adrenoceptor agonists, forskolin, PGE(2), 8-bromo-cAMP, or rolipram. Collectively, these results indicate that repression of IL-1beta-induced ICAM-1 expression and GM-CSF release by cAMP-elevating agents, including beta(2)-adrenoceptor agonists, may not occur through a generic effect on NF-kappaB.
Collapse
Affiliation(s)
- Manminder Kaur
- Dept. of Cell Biology & Anatomy, Faculty of Medicine, Univ. of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada T2N 4N1
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Pelaia G, Renda T, Gallelli L, Vatrella A, Busceti MT, Agati S, Caputi M, Cazzola M, Maselli R, Marsico SA. Molecular mechanisms underlying airway smooth muscle contraction and proliferation: implications for asthma. Respir Med 2008; 102:1173-81. [PMID: 18579364 DOI: 10.1016/j.rmed.2008.02.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 02/26/2008] [Indexed: 11/18/2022]
Abstract
Airway smooth muscle (ASM) plays a key role in bronchomotor tone, as well as in structural remodeling of the bronchial wall. Therefore, ASM contraction and proliferation significantly participate in the development and progression of asthma. Many contractile agonists also behave as mitogenic stimuli, thus contributing to frame a hyperresponsive and hyperplastic ASM phenotype. In this review, the molecular mechanisms and signaling pathways involved in excitation-contraction coupling and ASM cell growth will be outlined. Indeed, the recent advances in understanding the basic aspects of ASM biology are disclosing important cellular targets, currently explored for the implementation of new, more effective anti-asthma therapies.
Collapse
Affiliation(s)
- Girolamo Pelaia
- Department of Experimental and Clinical Medicine, University "Magna Graecia" of Catanzaro, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Qiao LF, Xu YJ, Liu XS, Xie JG, Wang J, Du CL, Zhang J, Ni W, Chen SX. PKC promotes proliferation of airway smooth muscle cells by regulating cyclinD1 expression in asthmatic rats. Acta Pharmacol Sin 2008; 29:677-86. [PMID: 18501114 PMCID: PMC7091861 DOI: 10.1111/j.1745-7254.2008.00795.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aim: To determine whether protein kinase C (PKC) has any effect on the expression of cyclinD1, a key regulator of growth control and G1/S transition, and to investigate the underlying molecular mechanisms of PKC involving the remodeling of the asthmatic airway smooth muscle (ASM). Methods: The treatment of synchronized ASM cells from asthmatic rats with PKC-specific agonist phorbol 12-myristate 13-acetate (PMA) and antagonist 2-{1-[3-(amidinothio) propyl]-1H-indol-3-yl}-3-(1-methylindol-3-yl) maleimide methanesulfonate salt (Ro31-8220) was followed by the proliferation assay. PKCα and cyclinD1 expressions in ASM cells (ASMC) were detected by RT-PCR and Western blotting. The relation between PKCα and cyclinD1 was assessed by linear regression analysis. The effect of the construct recombinant plasmid pcDNA3.1-antisense cyclinD1 (pcDNA3.1-ascyclinD1) on the proliferation of ASMC was found to be induced by PMA. Results: The data showed phorbol ester-dependent PKCα promoted the proliferation of ASMC. The closely-positive correlation existed between the expression of PKCα and cyclinD1 at the transcriptional (r=0.821, P<0.01) and transla-tional (r=0.940, P<0.01) levels. pcDNA3.1-ascyclinD1 could inhibit the proliferation of ASMC. pcDNA3.1-ascyclinD1 almost completely attenuated the PMA-induced proliferation effect as Ro31-8220+pcDNA3.1. Conclusion: The proliferation of ASMC by PKC might by regulated by the cyclinD1 expression in asthmatic rats.
Collapse
Affiliation(s)
- Li-fen Qiao
- Department of Respiratory Medicine, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Yong-jian Xu
- Department of Respiratory Medicine, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Xian-sheng Liu
- Department of Respiratory Medicine, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Jun-gang Xie
- Department of Respiratory Medicine, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Jin Wang
- Department of Emergency Medicine, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Chun-ling Du
- Department of Respiratory Medicine, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Jian Zhang
- Department of Respiratory Medicine, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Wang Ni
- Department of Respiratory Medicine, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Shi-xin Chen
- Department of Respiratory Medicine, Huazhong University of Science and Technology, Wuhan, 430030 China
| |
Collapse
|
28
|
Regulation of heterotrimeric G protein signaling in airway smooth muscle. Ann Am Thorac Soc 2008; 5:47-57. [PMID: 18094084 DOI: 10.1513/pats.200705-054vs] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Heterotrimeric G proteins transduce signals from G protein-coupled receptors to regulate numerous signaling events and functions in airway smooth muscle (ASM). In this article, we detail the function and regulation of heterotrimeric G protein signaling in ASM. We further discuss recent advances in the development of experimental tools in the study of G protein signaling, and speculate how these tools might be used in therapeutic strategies that seek to mitigate bronchospasm and airway remodeling that occur in obstructive airway disease.
Collapse
|
29
|
Geronikaki AA, Lagunin AA, Hadjipavlou-Litina DI, Eleftheriou PT, Filimonov DA, Poroikov VV, Alam I, Saxena AK. Computer-Aided Discovery of Anti-Inflammatory Thiazolidinones with Dual Cyclooxygenase/Lipoxygenase Inhibition. J Med Chem 2008; 51:1601-9. [DOI: 10.1021/jm701496h] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
30
|
Misior AM, Yan H, Pascual RM, Deshpande DA, Panettieri RA, Penn RB. Mitogenic Effects of Cytokines on Smooth Muscle Are Critically Dependent on Protein Kinase A and Are Unmasked by Steroids and Cyclooxygenase Inhibitors. Mol Pharmacol 2007; 73:566-74. [DOI: 10.1124/mol.107.040519] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
31
|
Petrovic N, Knight DA, Bomalaski JS, Thompson PJ, Misso NLA. Concomitant activation of extracellular signal-regulated kinase and induction of COX-2 stimulates maximum prostaglandin E2 synthesis in human airway epithelial cells. Prostaglandins Other Lipid Mediat 2006; 81:126-35. [PMID: 17085321 DOI: 10.1016/j.prostaglandins.2006.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2006] [Revised: 08/01/2006] [Accepted: 08/30/2006] [Indexed: 11/26/2022]
Abstract
The intracellular regulation and kinetics of prostaglandin (PG)E(2) synthesis in human airway epithelial (NCI-H292) cells was investigated. Interleukin (IL)-1beta, tumor necrosis factor (TNF)-alpha and lipopolysaccharide (LPS) all induced PGE(2) synthesis (p<0.001) and transient (5-15 min) phosphorylation of extracellular signal-regulated kinase (ERK). Phorbol myristate acetate (PMA) and calcium ionophore, A23187 further enhanced PGE(2) synthesis (p<0.001) and caused phosphorylation of ERK that was sustained for up to 16 h. COX-2 protein expression and PGE(2) synthesis were increased following exposure to combinations of stimuli that increased intracellular Ca(2+), and activated protein kinase C as well as ERK. Inhibition of ERK almost completely abrogated PGE(2) synthesis in response to all stimuli. Sustained, maximum PGE(2) synthesis was observed when cells were stimulated such that ERK phosphorylation was concomitant with increased COX-2 protein expression. These results argue against redundancy in pathways for PGE(2) synthesis, and suggest that at various stages of inflammation different stimuli may influence ERK activation and COX-2 expression, so as to tightly regulate the kinetics and amount of PGE(2) produced by airway epithelial cells in response to lung inflammation.
Collapse
Affiliation(s)
- Nenad Petrovic
- Lung Institute of Western Australia and Centre for Asthma, Allergy and Respiratory Research, The University of Western Australia, Perth, Australia
| | | | | | | | | |
Collapse
|
32
|
Abstract
The airway smooth muscle is the key determinant of airway narrowing in asthma but its function in the absence of disease is unknown. Evidence for an intrinsic abnormality in the muscle in asthma is only just emerging. The airway smooth muscle is not merely a contractile cell, but also one which determines the composition of, and interacts with the extracellular matrix, and which may participate in inflammatory and allergic reactions and viral infections. The reason for the differences which have been observed in the in vitro properties of airway smooth muscle derived from asthmatic individuals may result from an inherent "supercontractility", an increased tendency to proliferate due to the absence of an inhibitory transcription factor C/EBP-alpha, the influence of an altered extracellular matrix and/or a decrease in release of factors such as PGE(2) which would under normal circumstances inhibit both proliferation and contraction. Although long acting beta agonists and corticosteroids are successful treatments for inflammation and bronchoconstriction, the structural changes which constitute airway remodelling may require additional therapeutic intervention, the nature of which will be determined by thorough investigation of the mechanisms underlying the asthmatic phenotype.
Collapse
Affiliation(s)
- Brian G Oliver
- School of Medical Sciences Pharmacology, University of Sydney, NSW, Australia
| | | |
Collapse
|
33
|
Deshpande DA, Penn RB. Targeting G protein-coupled receptor signaling in asthma. Cell Signal 2006; 18:2105-20. [PMID: 16828259 DOI: 10.1016/j.cellsig.2006.04.008] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2006] [Accepted: 04/28/2006] [Indexed: 01/23/2023]
Abstract
The complex disease asthma, an obstructive lung disease in which excessive airway smooth muscle (ASM) contraction as well as increased ASM mass reduces airway lumen size and limits airflow, can be viewed as a consequence of aberrant airway G protein-coupled receptor (GPCR) function. The central role of GPCRs in determining airway resistance is underscored by the fact that almost every drug used in the treatment of asthma directly or indirectly targets either GPCR-ligand interaction, GPCR signaling, or processes that produce GPCR agonists. Although many airway cells contribute to the regulation of airway resistance and architecture, ASM properties and functions have the greatest impact on airway homeostasis. The theme of this review is that GPCR-mediated regulation of ASM tone and ASM growth is a major determinant of the acute and chronic features of asthma, and multiple strategies targeting GPCR signaling may be employed to prevent or manage these features.
Collapse
Affiliation(s)
- Deepak A Deshpande
- Department of Internal Medicine and Center for Human Genomics, Wake Forest University Health Sciences, Medical Center Blvd, Winston-Salem, NC 27157, United States
| | | |
Collapse
|
34
|
Lazaar AL, Panettieri RA. Airway smooth muscle as a regulator of immune responses and bronchomotor tone. Clin Chest Med 2006; 27:53-69, vi. [PMID: 16543052 DOI: 10.1016/j.ccm.2005.10.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The traditional view of airway smooth muscle (ASM) in asthma, as a purely contractile tissue, seems to be inadequate. Compelling evidence now suggests that ASM plays an important role in regulating bronchomotor tone, in perpetuating airway inflammation, and in remodeling of the airways. This article reviews three distinct functions of ASM cells: the process of excitation-contraction coupling, with a particular focus on the role of cytokines in modulating calcium responses; the processes of smooth muscle cell proliferation and migration; and the synthetic and immunomodulatory function of ASM cells. This article also discusses how altered synthetic function contributes to airway remodeling.
Collapse
Affiliation(s)
- Aili L Lazaar
- Pulmonary, Allergy, and Critical Care Division, University of Pennsylvania Medical Center, BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104-6160, USA.
| | | |
Collapse
|
35
|
Pascual RM, Carr EM, Seeds MC, Guo M, Panettieri RA, Peters SP, Penn RB. Regulatory features of interleukin-1β-mediated prostaglandin E2 synthesis in airway smooth muscle. Am J Physiol Lung Cell Mol Physiol 2006; 290:L501-8. [PMID: 16299051 DOI: 10.1152/ajplung.00420.2005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exposure of airway smooth muscle (ASM) cells to the cytokine IL-1β results in an induction of PGE2 synthesis that affects numerous cell functions. Current dogma posits induction of COX-2 protein as the critical, obligatory event in cytokine-induced PGE2 production, although PGE2 induction can be inhibited without a concomitant inhibition of COX-2. To explore other putative regulatory features we examined the role of phospholipase A2 (PLA2) and PGE synthase (PGES) enzymes in IL-1β-induced PGE2 production. Treatment of human ASM cultures with IL-1β caused a time-dependent induction of both cytosolic PLA2 (cPLA2) and microsomal PGES (mPGES) similar to that observed for COX-2. Regulation of COX-2 and mPGES induction was similar, being significantly reduced by inhibition of p42/p44 or p38, whereas cPLA2 induction was only minimally reduced by inhibition of p38 or PKC. COX-2 and mPGES induction was subject to feed-forward regulation by PKA, whereas cPLA2 induction was not. SB-202474, an SB-203580 analog lacking the ability to inhibit p38 but capable of inhibiting IL-1β-induced PGE2 production, was effective in inhibiting mPGES but not COX-2 or cPLA2 induction. These data suggest that although COX-2, cPLA2, and mPGES are all induced by IL-β in human ASM cells, regulatory features of cPLA2 are dissociated, whereas those of COX-2 and mPGES are primarily associated, with regulation of PGE2 production. mPGES induction and, possibly, cPLA2 induction appear to cooperate with COX-2 to determine IL-1β-mediated PGE2 production in human ASM cells.
Collapse
Affiliation(s)
- Rodolfo M Pascual
- Department of Internal Medicine and Center for Human Genomics, Wake Forest Univ. Health Sciences Center, Center for Human Genomics, Medical Center Blvd, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Kothapalli D, Flores-Stewart SA, Assoian RK. Antimitogenic effects of prostacyclin on the G1 phase cyclin-dependent kinases. Prostaglandins Other Lipid Mediat 2005; 78:3-13. [PMID: 16303599 DOI: 10.1016/j.prostaglandins.2005.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2005] [Accepted: 04/01/2005] [Indexed: 12/12/2022]
Abstract
The prostanoid prostacyclin (PGI2) inhibits proliferation of cultured vascular SMCs by inhibiting cell cycle progression from G1 to S phase. Progression through G1 phase is regulated by the sequential activation of the G1 phase cyclin-dependent kinases (cdks). Recent studies have shown that PGI2-dependent activation of its receptor, IP, inhibits G1 phase progression by blocking the degradation of p27 and the activation of cyclin E-cdk2. High Density Lipoproteins (HDL) and its associated apolipoprotein, ApoE, also inhibit S phase entry of vascular SMCs, and the effects of HDL and ApoE are, at least in part, also mediated by the production of PGI2. The antimitogenic effects of hyaluronan may also be controlled by PGI2. This review summarizes the effects of PGI2 on the G1 phase cyclin-cdks and discusses the potential role of PGI2 as a common component of multiple extracellular signals that attenuate the proliferation of vascular SMCs.
Collapse
Affiliation(s)
- Devashish Kothapalli
- Department of Pharmacology, University of Pennsylvania School of Medicine, 3620 Hamilton Walk, 167 Johnson Pavilion, Philadelphia, PA 19104-6084, USA
| | | | | |
Collapse
|
37
|
Guo M, Pascual RM, Wang S, Fontana MF, Valancius CA, Panettieri RA, Tilley SL, Penn RB. Cytokines regulate beta-2-adrenergic receptor responsiveness in airway smooth muscle via multiple PKA- and EP2 receptor-dependent mechanisms. Biochemistry 2005; 44:13771-82. [PMID: 16229467 DOI: 10.1021/bi051255y] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Beta2AR desensitization in airway smooth muscle (ASM) mediated by airway inflammation has been proposed to contribute to asthma pathogenesis and diminished efficacy of beta-agonist therapy. Mechanistic insight into this phenomenon is largely conceptual and lacks direct empirical evidence. Here, we employ molecular and genetic strategies to reveal mechanisms mediating cytokine effects on ASM beta2AR responsiveness. Ectopic expression of inhibitory peptide (PKI-GFP) or a mutant regulatory subunit of PKA (RevAB-GFP) effectively inhibited intracellular PKA activity in cultured human ASM cells and enhanced beta2AR responsiveness by mitigating both agonist-specific (beta-agonist-mediated) desensitization and cytokine (IL-1beta and TNF-alpha)-induced heterologous desensitization via actions on multiple targets. In the absence of cytokine treatment, PKA inhibition increased beta2AR-mediated signaling by increasing both beta2AR-G protein coupling and intrinsic adenylyl cyclase activity. PKI-GFP and RevAB-GFP expression also conferred resistance to cytokine-promoted beta2AR-G protein uncoupling and disrupted feed-forward mechanisms of PKA activation by attenuating the induction of COX-2 and PGE2. Cytokine treatment of tracheal ring preparations from wild-type mice resulted in a profound loss of beta-agonist-mediated relaxation of methacholine-contracted rings, whereas rings from EP2 receptor knockout mice were largely resistant to cytokine-mediated beta2AR desensitization. These findings identify EP2 receptor- and PKA-dependent mechanisms as the principal effectors of cytokine-mediated beta2AR desensitization in ASM.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cells, Cultured
- Cytokines/pharmacology
- Enzyme Activation
- Green Fluorescent Proteins/metabolism
- Humans
- In Vitro Techniques
- Mice
- Muscle, Smooth/cytology
- Muscle, Smooth/drug effects
- Muscle, Smooth/enzymology
- Muscle, Smooth/metabolism
- Protein Kinase C/antagonists & inhibitors
- Protein Kinase C/metabolism
- Receptors, Adrenergic, alpha-2/physiology
- Receptors, Prostaglandin E/metabolism
- Receptors, Prostaglandin E, EP2 Subtype
- Trachea/cytology
- Trachea/drug effects
- Trachea/enzymology
- Trachea/metabolism
Collapse
Affiliation(s)
- Manhong Guo
- Department of Internal Medicine and Center for Human Genomics, Wake Forest University Health Sciences Center, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Heterotrimeric G proteins are key players in transmembrane signaling by coupling a huge variety of receptors to channel proteins, enzymes, and other effector molecules. Multiple subforms of G proteins together with receptors, effectors, and various regulatory proteins represent the components of a highly versatile signal transduction system. G protein-mediated signaling is employed by virtually all cells in the mammalian organism and is centrally involved in diverse physiological functions such as perception of sensory information, modulation of synaptic transmission, hormone release and actions, regulation of cell contraction and migration, or cell growth and differentiation. In this review, some of the functions of heterotrimeric G proteins in defined cells and tissues are described.
Collapse
Affiliation(s)
- Nina Wettschureck
- Institute of Pharmacology, University of Heidelberg, Im Neuenheimer Feld 366, D-69120 Heidelberg, Germany
| | | |
Collapse
|
39
|
Burgess JK, Blake AE, Boustany S, Johnson PRA, Armour CL, Black JL, Hunt NH, Hughes JM. CD40 and OX40 ligand are increased on stimulated asthmatic airway smooth muscle. J Allergy Clin Immunol 2005; 115:302-8. [PMID: 15696085 DOI: 10.1016/j.jaci.2004.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Severe, persistent asthma is characterized by airway smooth muscle hyperplasia, inflammatory cell infiltration into the smooth muscle, and increased expression of many cytokines, including IL-4, IL-13, IL-1beta, and TNF-alpha. These cytokines have the potential to alter the expression of surface receptors such as CD40 and OX40 ligand on the airway smooth muscle cell. OBJECTIVE To examine whether cytokines alter expression of CD40 and OX40 ligand on airway smooth muscle cells and identify any differences in response between asthmatic and nonasthmatic airway smooth muscle cells. METHODS We used flow cytometry and immunohistochemistry to detect CD40 and OX40 ligand on airway smooth muscle cells cultured in the presence of TNF-alpha, IL-1beta, IL-4, or IL-13. Prostaglandin E 2 levels were assessed by ELISA. RESULTS TNF-alpha increased expression of both CD40 and OX40 ligand on both asthmatic and nonasthmatic airway smooth muscle cells. The level of expression was significantly greater on the asthmatic cells. IL-1beta alone had no effect, but it attenuated the TNF-induced expression of both CD40 and OX40 ligand. The mechanism of inhibition was COX-dependent for CD40 and was COX-independent but cyclic AMP-dependent for OX40 ligand. IL-4 and IL-13 had no effect. CONCLUSION Our study has demonstrated that TNF-alpha and IL-1beta have the potential to modulate differentially the interactions between cells present in the inflamed airways of a patient with asthma and therefore to contribute to the regulation of airway inflammation and remodeling.
Collapse
Affiliation(s)
- Janette K Burgess
- Department of Pharmacology, University of Sydney, Sydney, NSW, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Affiliation(s)
- Marc Peters-Golden
- Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109-0642, USA.
| |
Collapse
|
41
|
Ritchie RH, Rosenkranz AC, Huynh LP, Stephenson T, Kaye DM, Dusting GJ. Activation of IP prostanoid receptors prevents cardiomyocyte hypertrophy via cAMP-dependent signaling. Am J Physiol Heart Circ Physiol 2004; 287:H1179-85. [PMID: 15072955 DOI: 10.1152/ajpheart.00725.2003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The antihypertrophic action of angiotensin-converting enzyme inhibitors in the heart results partly from local potentiation of bradykinin. We have demonstrated that the antihypertrophic action of bradykinin is mediated by the release of nitric oxide from endothelium and elevation of cardiomyocyte cGMP. Whether other paracrine factors derived from the coronary endothelium, such as prostacyclin (PGI2), may act to prevent hypertrophy has not been explored. In the vasculature, activation by PGI2 of IP and EP1 prostanoid receptors elicits vasodilatation (via cAMP-dependent signaling) and vasoconstriction, respectively. The present objective was to determine whether IP prostanoid receptor activation has antihypertrophic actions in adult rat cardiomyocytes (ARCM). The selective IP agonist cicaprost (1 microM) virtually abolished the increase in [3H]phenylalanine incorporation (a marker of hypertrophy) induced either by endothelin-1 (ET-1; 60 nM, n = 10, P < 0.005) or by angiotensin II (1 microM, n = 6, P < 0.005). Cicaprost also inhibited ET-1 induction of c-fos mRNA expression, an additional marker of hypertrophy in ARCM (n = 5, P < 0.005). In the absence of hypertrophic stimuli, cicaprost alone did not significantly influence either marker. The antihypertrophic actions of cicaprost were mimicked by the dual IP/EP1 agonist iloprost (1 microM) in the presence of the EP1 antagonist AH-6809 (3 microM). Furthermore, cicaprost modestly but significantly increased cardiomyocyte cAMP content by 13 +/- 6% (P < 0.05, n = 4), and the antihypertrophic effect of cicaprost was lost in the presence of the cAMP-dependent protein kinase inhibitor H-89 (1 microM, n = 5, P < 0.05). However, ET-1 also induced increases in the activity of the intracellular growth signals ERK1 (by 3-fold) and ERK2 (by 5-fold) in ARCM, and these were not inhibited by cicaprost (P < 0.01, n = 5). Activation of IP receptors thus represents a novel approach to prevention of hypertrophy, and this effect is linked to cAMP-dependent signaling.
Collapse
|
42
|
Mitchell BF, Olson DM. Prostaglandin endoperoxide H synthase inhibitors and other tocolytics in preterm labour. Prostaglandins Leukot Essent Fatty Acids 2004; 70:167-87. [PMID: 14683691 DOI: 10.1016/j.plefa.2003.04.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Preterm delivery (<37 weeks of gestation) is the major obstetrical complication in developed countries, yet attempts to delay labour and prolong pregnancy have largely been unsuccessful. One of the many reasons it is so difficult to prevent preterm birth is that the nature of preterm labour changes as a function of gestational age, maternal lifestyle factors or infection, to list a few of the reasons. The inhibitors of prostaglandin endoperoxide H synthase (PGHS), known as the Non-steroidal Antiinflammatory Drugs, have been viewed with interest as tocolytics with promising effectiveness under most conditions of preterm labour. Three isoforms of PGHS exist; the first two, PGHS-1 and -2, have been studied for their catalytic activity, X-ray crystallographic structure, and physiological roles in the adult and the foetus. Mixed inhibitors and isoform-specific inhibitors of PGHS have been developed, and their roles in delaying preterm labour are examined and compared to other tocolytics.
Collapse
Affiliation(s)
- Bryan F Mitchell
- Department of Obstetrics and Gynaecology, Perinatal Research Centre, CIHR Group in Perinatal Health and Disease, University of Alberta, 220 HMRC, Edmonton, Alberta, Canada T6G2S2
| | | |
Collapse
|
43
|
Nie M, Pang L, Inoue H, Knox AJ. Transcriptional regulation of cyclooxygenase 2 by bradykinin and interleukin-1beta in human airway smooth muscle cells: involvement of different promoter elements, transcription factors, and histone h4 acetylation. Mol Cell Biol 2004; 23:9233-44. [PMID: 14645533 PMCID: PMC309638 DOI: 10.1128/mcb.23.24.9233-9244.2003] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bradykinin and interleukin-1beta (IL-1beta) induce cyclooxygenase 2 (COX-2) in human airway smooth muscle cells. Here we extended our study to explore the gene transcriptional regulation. By transfection with various COX-2 promoter reporter constructs, we found that the bp -327-to-+59 promoter region was essential for COX-2 gene transcription by bradykinin and IL-1beta and that the cyclic AMP response element (CRE) was critical in bradykinin-induced transcription, whereas nuclear factor IL-6 and CRE and, to a lesser extent, nuclear factor-kappaB (NF-kappaB) were involved in IL-1beta-induced transcription. An electrophoretic mobility shift assay revealed that both bradykinin and IL-1beta elicited CRE-binding protein-1 (CREB-1) binding, and IL-1beta also elicited CCAAT/enhancer-binding protein beta and NF-kappaB binding to their respective elements in the COX-2 promoter. These transcription factors were associated with the COX-2 promoter, which was dynamically linked to different patterns of histone H4 acetylation by bradykinin and IL-1beta, as demonstrated by chromatin immunoprecipitation. We also revealed that endogenous prostaglandin E(2) was critical in bradykinin-induced COX-2 transcription initiation and involved in IL-1beta-induced COX-2 transcription at a latter stage. Our result provide the first evidence that COX-2 transcriptional regulation by different stimuli involves different promoter elements and transcription factors and is associated with chromatin remodeling after selective histone H4 acetylation in a stimulus-specific manner.
Collapse
Affiliation(s)
- Mei Nie
- Division of Respiratory Medicine, City Hospital, University of Nottingham, Hucknall Road, Nottingham NG5 1PB, United Kingdom.
| | | | | | | |
Collapse
|
44
|
Panettieri RA. Airway smooth muscle: immunomodulatory cells that modulate airway remodeling? Respir Physiol Neurobiol 2003; 137:277-93. [PMID: 14516732 DOI: 10.1016/s1569-9048(03)00153-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Although the pathogenesis of asthma remains unclear, substantial progress has been made over the past decades in the characterization of airway inflammation as a pathogenetic mechanism in asthma. New evidence suggests that airway smooth muscle (ASM), the most important cell modulating bronchomotor tone, plays an important immunomodulatory role in the orchestration and perpetuation of airway inflammation. Evidence now suggests that the signaling pathways that modulate leukocyte function may be disparate from those found in resident effector cells such as ASM, fibroblasts and epithelial cells. Further investigation and understanding of the critical signaling pathways that modulate ASM cell release, secretion of chemokines/cytokines and expression of cell adhesion molecules (CAMs) may offer new therapeutic approaches in the treatment of asthma.
Collapse
Affiliation(s)
- Reynold A Panettieri
- Pulmonary, Allergy and Critical Care Division, University of Pennsylvania Medical Center, 421 Curie Boulevard, 805 BRB II/III, Philadelphia, Pennsylvania, PA 19104-6160, USA.
| |
Collapse
|
45
|
Chambers LS, Black JL, Ge Q, Carlin SM, Au WW, Poniris M, Thompson J, Johnson PR, Burgess JK. PAR-2 activation, PGE2, and COX-2 in human asthmatic and nonasthmatic airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2003; 285:L619-27. [PMID: 12754192 DOI: 10.1152/ajplung.00416.2002] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The protease-activated receptor-2 (PAR-2) is present on human airway smooth muscle (ASM) cells and can be activated by mast cell tryptase, trypsin, or an activating peptide (AP). Trypsin induced significant increases in PGE2 release from human ASM cells after 6 and 24 h and also induced cyclooxygenase (COX)-2 mRNA expression and COX-2 protein. Tryptase and the PAR-2 AP did not alter PGE2 release or COX-2 protein levels, suggesting a lack of PAR-2 involvement. When we compared results in asthmatic and nonasthmatic muscle cells, both trypsin and bradykinin induced less PGE2 from asthmatic ASM cells, and bradykinin induced significantly less COX-2 mRNA in asthmatic cells. Significantly less PGE2 was released from proliferating ASM cells from asthmatic patients. In conclusion, trypsin induces PGE2 release and COX-2 in human ASM cells, which is unlikely to be via PAR-2 activation. In addition, ASM cells from asthmatic patients produce significantly less PGE2 and COX-2 compared with nonasthmatic cells. These findings may contribute to the increase in muscle mass evident in asthmatic airways.
Collapse
Affiliation(s)
- Linda S Chambers
- Dept. of Pharmacology, Univ. of Sydney, New South Wales 2006, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ballaz S, Mulshine JL. The Potential Contributions of Chronic Inflammation to Lung Carcinogenesis. Clin Lung Cancer 2003; 5:46-62. [PMID: 14596704 DOI: 10.3816/clc.2003.n.021] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A number of lines of evidence suggests that chronic inflammation contributes to the process of carcinogenesis. In this article, this theme is explored with particular emphasis on the involvement of inflammation in the development of lung cancer. A number of molecular pathways activated in chronic inflammation may contribute to lung carcinogenesis. The challenge is to conceptualize a cohesive picture of this complex biology that allows for effective pharmaceutical intervention. Initial therapeutic efforts involve strategies to block single pathways, such as with cyclooxygenase (COX) activity. However, the more that is learned about the consequences of COX activity, the more evident are the relationships of this enzyme to other classes of regulatory molecules such as the potent nuclear factor-kB. In light of this emerging picture, more global intervention strategies, such as with drug combinations, may be essential for success. Further basic study is essential to sort out possible molecular relationships and to permit elucidation of the most critical regulatory circuits. Given the complexity of these molecular interactions, well-designed clinical trials that specifically evaluate the precise effects of particular antiinflammatory drugs on lung carcinogenesis will also be critical to sort out the complexity and to validate successful approaches to arresting lung carcinogenesis.
Collapse
Affiliation(s)
- Santiago Ballaz
- Department of Histology and Pathology, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | |
Collapse
|
47
|
Martel-Pelletier J, Lajeunesse D, Reboul P, Pelletier JP. Therapeutic role of dual inhibitors of 5-LOX and COX, selective and non-selective non-steroidal anti-inflammatory drugs. Ann Rheum Dis 2003; 62:501-9. [PMID: 12759283 PMCID: PMC1754580 DOI: 10.1136/ard.62.6.501] [Citation(s) in RCA: 300] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Dual 5-LOX/COX inhibitors are potential new drugs to treat inflammation. They act by blocking the formation of both prostaglandins and leucotrienes but do not affect lipoxin formation. Such combined inhibition avoids some of the disadvantages of selective COX-2 inhibitors and spares the gatrointestinal mucosa.
Collapse
Affiliation(s)
- J Martel-Pelletier
- Osteoarthritis Research Unit, Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, Montréal, Québec H2L 4M1 Canada.
| | | | | | | |
Collapse
|
48
|
Billington CK, Penn RB. Signaling and regulation of G protein-coupled receptors in airway smooth muscle. Respir Res 2003. [DOI: 10.1186/1465-9921-4-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
49
|
Billington CK, Penn RB. Signaling and regulation of G protein-coupled receptors in airway smooth muscle. Respir Res 2003; 4:2. [PMID: 12648290 PMCID: PMC152647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2002] [Revised: 10/09/2002] [Accepted: 10/14/2002] [Indexed: 11/27/2022] Open
Abstract
Signaling through G protein-coupled receptors (GPCRs) mediates numerous airway smooth muscle (ASM) functions including contraction, growth, and "synthetic" functions that orchestrate airway inflammation and promote remodeling of airway architecture. In this review we provide a comprehensive overview of the GPCRs that have been identified in ASM cells, and discuss the extent to which signaling via these GPCRs has been characterized and linked to distinct ASM functions. In addition, we examine the role of GPCR signaling and its regulation in asthma and asthma treatment, and suggest an integrative model whereby an imbalance of GPCR-derived signals in ASM cells contributes to the asthmatic state.
Collapse
Affiliation(s)
- Charlotte K Billington
- Department of Medicine, Division of Critical Care, Pulmonary, Allergic & Immunologic Diseases, and Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| | - Raymond B Penn
- Department of Medicine, Division of Critical Care, Pulmonary, Allergic & Immunologic Diseases, and Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
50
|
Patel HJ, Belvisi MG, Bishop-Bailey D, Yacoub MH, Mitchell JA. Activation of peroxisome proliferator-activated receptors in human airway smooth muscle cells has a superior anti-inflammatory profile to corticosteroids: relevance for chronic obstructive pulmonary disease therapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:2663-9. [PMID: 12594295 DOI: 10.4049/jimmunol.170.5.2663] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Airway smooth muscle is actively involved in the inflammatory process in diseases such as chronic obstructive pulmonary disease and asthma by 1) contributing to airway narrowing through hyperplasia and hypertrophy and 2) the release of GM-CSF and G-CSF, which promotes the survival and activation of infiltrating leukocytes. Thus, the identification of novel anti-inflammatory pathways in airway smooth muscle will have important implications for the treatment of inflammatory airway disease. This study identifies such a pathway in the activation of peroxisome proliferator-activated receptors (PPARs). PPAR ligands are known therapeutic agents in the treatment of diabetes; however, their role in human airway disease is unknown. We demonstrate, for the first time, that human airway smooth muscle cells express PPAR alpha and -gamma subtypes. Activation of PPAR gamma by natural and synthetic ligands inhibits serum-induced cell growth more effectively than does the steroid dexamethasone, and induces apoptosis. Moreover, PPAR gamma activation, like dexamethasone, inhibits the release of GM-CSF. However, PPAR gamma ligands, but not dexamethasone, similarly inhibits G-CSF release. These results reveal a novel anti-inflammatory pathway in human airway smooth muscle, where PPAR gamma activation has additional anti-inflammatory effects to those of steroids. Hence, PPAR ligands might act as potential treatments in human respiratory diseases.
Collapse
MESH Headings
- Adolescent
- Adult
- Anti-Inflammatory Agents/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- Cell Division/drug effects
- Cells, Cultured
- DNA Fragmentation/drug effects
- Dexamethasone/pharmacology
- Female
- Granulocyte Colony-Stimulating Factor/metabolism
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Growth Inhibitors/pharmacology
- Humans
- Interleukin-1/pharmacology
- Ligands
- Male
- Middle Aged
- Muscle, Smooth/cytology
- Muscle, Smooth/metabolism
- Peroxisomes/genetics
- Peroxisomes/metabolism
- Prostaglandin D2/analogs & derivatives
- Prostaglandin D2/metabolism
- Prostaglandin D2/pharmacology
- Protein Isoforms/biosynthesis
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- Pulmonary Disease, Chronic Obstructive/drug therapy
- Pulmonary Disease, Chronic Obstructive/therapy
- Pyrimidines/metabolism
- Pyrimidines/pharmacology
- RNA, Messenger/biosynthesis
- Receptors, Cytoplasmic and Nuclear/biosynthesis
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Thiazoles/metabolism
- Thiazoles/pharmacology
- Thiazolidinediones
- Trachea/cytology
- Trachea/metabolism
- Transcription Factors/biosynthesis
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Hema J Patel
- Respiratory Pharmacology Group, Department of Cardiothoracic Surgery, Faculty of Medicine, The National Heart and Lung Institute, Imperial College School of Medicine, London, United Kingdom.
| | | | | | | | | |
Collapse
|