1
|
Mokra D, Mokry J. Phosphodiesterase Inhibitors in Acute Lung Injury: What Are the Perspectives? Int J Mol Sci 2021; 22:1929. [PMID: 33669167 PMCID: PMC7919656 DOI: 10.3390/ijms22041929] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/14/2022] Open
Abstract
Despite progress in understanding the pathophysiology of acute lung damage, currently approved treatment possibilities are limited to lung-protective ventilation, prone positioning, and supportive interventions. Various pharmacological approaches have also been tested, with neuromuscular blockers and corticosteroids considered as the most promising. However, inhibitors of phosphodiesterases (PDEs) also exert a broad spectrum of favorable effects potentially beneficial in acute lung damage. This article reviews pharmacological action and therapeutical potential of nonselective and selective PDE inhibitors and summarizes the results from available studies focused on the use of PDE inhibitors in animal models and clinical studies, including their adverse effects. The data suggest that xanthines as representatives of nonselective PDE inhibitors may reduce acute lung damage, and decrease mortality and length of hospital stay. Various (selective) PDE3, PDE4, and PDE5 inhibitors have also demonstrated stabilization of the pulmonary epithelial-endothelial barrier and reduction the sepsis- and inflammation-increased microvascular permeability, and suppression of the production of inflammatory mediators, which finally resulted in improved oxygenation and ventilatory parameters. However, the current lack of sufficient clinical evidence limits their recommendation for a broader use. A separate chapter focuses on involvement of cyclic adenosine monophosphate (cAMP) and PDE-related changes in its metabolism in association with coronavirus disease 2019 (COVID-19). The chapter illuminates perspectives of the use of PDE inhibitors as an add-on treatment based on actual experimental and clinical trials with preliminary data suggesting their potential benefit.
Collapse
Affiliation(s)
- Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Juraj Mokry
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia;
| |
Collapse
|
2
|
Yu HP, Liu FC, Lin CY, Umoro A, Trousil J, Hwang TL, Fang JY. Suppression of neutrophilic inflammation can be modulated by the droplet size of anti-inflammatory nanoemulsions. Nanomedicine (Lond) 2020; 15:773-791. [PMID: 32193978 DOI: 10.2217/nnm-2019-0407] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Aim: We aimed to develop nanoemulsions containing phosphodiesterase 4 inhibitor rolipram with different droplet sizes, to evaluate the anti-inflammatory effect against activated neutrophils and a related lung injury. Materials & methods: We prepared nanoemulsions of three different sizes, 68, 133 and 188 nm. Results: The nanoemulsion inhibited the superoxide anion but not elastase release in primary human neutrophils. The large-sized nanoemulsions were mostly internalized by neutrophils, resulting in the reduction of intracellular Ca2+ half-life. The peripheral organ distribution of near-infrared dye-tagged nanoemulsions increased, following the decrease in droplet diameter. Rolipram entrapment into intravenous nanoemulsions ameliorated pulmonary inflammation. The smallest droplet size showed improvement, compared with the largest size. Conclusion: We established a foundation for the development of nanoemulsions against inflamed lung disease.
Collapse
Affiliation(s)
- Huang-Ping Yu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, 333, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, 333, Taiwan.,School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan
| | - Cheng-Yu Lin
- Graduate Institute of Biomedical Sciences, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan
| | - Ani Umoro
- Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan
| | - Jiří Trousil
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Prague, 11720, Czech Republic
| | - Tsong-Long Hwang
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, 333, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan.,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan.,Research Center for Food & Cosmetic Safety & Research Center for Chinese Herbal Medicine, Chang Gung University of Science & Technology, Kweishan, Taoyuan, 333, Taiwan.,Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, 243, Taiwan
| | - Jia-You Fang
- Department of Anesthesiology, Chang Gung Memorial Hospital, Kweishan, Taoyuan, 333, Taiwan.,Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan.,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan, 333, Taiwan.,Research Center for Food & Cosmetic Safety & Research Center for Chinese Herbal Medicine, Chang Gung University of Science & Technology, Kweishan, Taoyuan, 333, Taiwan
| |
Collapse
|
3
|
Dillard J, Perez M, Chen B. Therapies that enhance pulmonary vascular NO-signaling in the neonate. Nitric Oxide 2019; 95:45-54. [PMID: 31870967 DOI: 10.1016/j.niox.2019.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 10/25/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023]
Abstract
There are several pulmonary hypertensive diseases that affect the neonatal population, including persistent pulmonary hypertension of the newborn (PPHN) and bronchopulmonary dysplasia (BPD)-associated pulmonary hypertension (PH). While the indication for inhaled nitric oxide (iNO) use is for late-preterm and term neonates with PPHN, there is a suboptimal response to this pulmonary vasodilator in ~40% of patients. Additionally, there are no FDA-approved treatments for BPD-associated PH or for preterm infants with PH. Therefore, investigating mechanisms that alter the nitric oxide-signaling pathway has been at the forefront of pulmonary vascular biology research. In this review, we will discuss the various mechanistic pathways that have been targets in neonatal PH, including NO precursors, soluble guanylate cyclase modulators, phosphodiesterase inhibitors and antioxidants. We will review their role in enhancing NO-signaling at the bench, in animal models, as well as highlight their role in the treatment of neonates with PH.
Collapse
Affiliation(s)
- Julie Dillard
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.
| | - Marta Perez
- Division of Neonatology, Stanley Manne Children's Research Institute, Ann and Robert H Lurie Children's Hospital, Chicago, IL, USA; Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Chicago, IL, USA.
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
4
|
Sharma V, Kumar H, Wakode S. Pharmacophore generation and atom based 3D-QSAR of quinoline derivatives as selective phosphodiesterase 4B inhibitors. RSC Adv 2016. [DOI: 10.1039/c6ra11210b] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Reported PDE4B inhibitors were used to design QSAR based pharmacophore model. Using developed pharmacophore model, virtual screening was performed followed by cross-docking to identify novel PDE4B specific inhibitors.
Collapse
Affiliation(s)
- Vidushi Sharma
- Department of Pharmaceutical Chemistry
- Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR)
- University of Delhi
- New Delhi – 110017
- India
| | - Hirdesh Kumar
- Parasitology – Center for Infectious Diseases
- University of Heidelberg Medical School
- 69120 Heidelberg
- Germany
| | - Sharad Wakode
- Department of Pharmaceutical Chemistry
- Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR)
- University of Delhi
- New Delhi – 110017
- India
| |
Collapse
|
5
|
Shirole RL, Shirole NL, Saraf MN. Embelia ribes ameliorates lipopolysaccharide-induced acute respiratory distress syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2015; 168:356-63. [PMID: 25818695 DOI: 10.1016/j.jep.2015.03.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/17/2015] [Accepted: 03/04/2015] [Indexed: 05/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Embelia ribes Burm. f. (Fam. Myrsinaceae) locally known as Vidanga have been used for treating tumors, ascites, bronchitis, jaundice, diseases of the heart and brain in traditional Indian medicine. However, no scientific studies providing new insights in its pharmacological properties with respect to acute respiratory distress syndrome have been investigated. AIM The present investigation aimed to elucidate the effectiveness of Embelin isolated from Embelia ribes seeds on attenuation of LPS-induced acute respiratory distress syndrome in murine models. METHODS Embelin (5, 10 and 20 mg/kg/day, i.p.) and Roflumilast (1 mg/kg/day, p.o.) were administered for four days and prior to LPS in rats (i.t.). Four hour after LPS challenge animals were anesthesized and bronchoalveolar lavage was done with ice-cold phosphate buffer. Assessment of BAL fluid was done for albumin, total protein, total cell and neutrophil count, TNF-α levels, nitrosoative stress. Superior lobe of right lung was used for histopathologic evaluation. Inferior lobe of right lung was used to obtain lung edema. Left lung was used for myeloperoxidase estimation. Arterial blood was collected immediately and analyzed for pH, pO2 and pCO2 were estimated. RESULTS Pretreatment with embelin (5, 10 and 20 mg/kg, i.p.) decreased lung edema, mononucleated cellular infiltration, nitrate/nitrite, total protein, albumin concentrations, TNF-α in the bronchoalveolar lavage fluid and myeloperoxidase activity in lung homogenate. Embelin markedly prevented pO2 down-regulation and pCO2 augmentation. Additionally, it attenuated lung histopathological changes in acute respiratory distress syndrome model. CONCLUSION The study demonstrates the effectiveness of Embelia ribes Burm. f. (Fam. Myrsinaceae) seeds in acute respiratory distress syndrome possibly related to its anti-inflammatory and protective effect against LPS induced airway inflammation by reducing nitrosative stress, reducing physiological parameters of blood gas change, TNF-α and mononucleated cellular infiltration indicating it as a potential therapeutic agent for acute respiratory distress syndrome.
Collapse
Affiliation(s)
- R L Shirole
- Department of Pharmacology, A. R. A. College of Pharmacy, Dhule, Maharashtra, India.
| | - N L Shirole
- Department of Pharmaceutical Chemistry, A. R. A. College of Pharmacy, Dhule, Maharashtra, India
| | - M N Saraf
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai, Maharashtra, India
| |
Collapse
|
6
|
Shirole RL, Shirole NL, Kshatriya AA, Kulkarni R, Saraf MN. Investigation into the mechanism of action of essential oil of Pistacia integerrima for its antiasthmatic activity. JOURNAL OF ETHNOPHARMACOLOGY 2014; 153:541-551. [PMID: 24556221 DOI: 10.1016/j.jep.2014.02.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 12/30/2013] [Accepted: 02/07/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pistacia integerrima J.L. Stewart ex Brandis locally known as Karkatashringi is an important medicinal plant whose galls are valued in traditional medicine used in India for the treatment of asthma, chronic bronchitis, phthisis, diarrhea, fever, other ailments for the respiratory tract, and as antispasmodic, carminative, antiamoebic and anthelmintic. However, in vitro and in vivo investigations providing new insights into its pharmacological properties have not been thoroughly investigated yet. The present investigation aimed to elucidate the probable mechanism of antiasthmatic action of essential oil of Pistacia integerrima J.L. Stewart ex Brandis galls (EOPI). METHODS EOPI was tested using in vitro studies such as antioxidant activity, mast cell degranulation, angiogenesis, isolated guinea pig ileum preparation and soyabean lipoxidase enzyme activity. In vivo studies included lipopolysaccharide-induced bronchial inflammation in rats and airway hyperresponsiveness in ovalbumin in sensitized guinea pigs using spirometry. RESULTS EOPI (5-30 µg/ml) inhibits 5-lipoxidase enzyme activity with IC50 of 19.71 µg/ml and DPPH scavenging activity up to 100 µg/ml with maximum inhibition of 44.93 ± 2.53% at 100 µg/ml. Pre-treatment with EOPI inhibited erythropoietin-induced angiogenesis. It showed dose dependent (10, 30 and 100 µg/ml) anti-allergic activity by inhibiting compound 48/80 induced mast cell degranulation to an extent 19.08 ± 0.47%. The finding that essential oil induced inhibition of transient contraction of acetylcholine in calcium free medium, and relaxation of S-(-)-Bay 8644-precontracted isolated guinea pig ileum jointly suggests that the L-subtype Cav channel is involved in spasmolytic action of EOPI. Treatment with EOPI dose dependently (7.5, 15 and 30mg/kg i.p.) inhibited lipopolysaccharide-induced increase in total cell count, neutrophil count, nitrate-nitrite, total protein, albumin levels in bronchoalveolar fluid and myeloperoxidase levels in lung homogenates. Roflumilast was used as a standard. EOPI reduced the respiratory flow due to gasping in ovalbumin sensitized guinea pigs. CONCLUSION The study demonstrates the effectiveness of essential oil of Pistacia integerrima J.L. Stewart ex Brandis galls in bronchial asthma possibly related to its ability to inhibit L-subtype Cav channel, mast cell stabilization, antioxidant, angiostatic and through inhibition of 5-lipoxygenase enzyme.
Collapse
Affiliation(s)
- R L Shirole
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai, India
| | - N L Shirole
- Department of Pharmaceutical Chemistry, A. R. A. College of Pharmacy, Nagaon, Dhule, India
| | - A A Kshatriya
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai, India
| | - R Kulkarni
- Department of Clinical Pharmacology T. N. Medical College and B. Y. L. Nair Charitable Hospital, Mumbai, India
| | - M N Saraf
- Department of Pharmacology, Bombay College of Pharmacy, Kalina, Santacruz (E), Mumbai, India.
| |
Collapse
|
7
|
Ting PC, Lee JF, Kuang R, Cao J, Gu D, Huang Y, Liu Z, Aslanian RG, Feng KI, Prelusky D, Lamca J, House A, Phillips JE, Wang P, Wu P, Lundell D, Chapman RW, Celly CS. Discovery of oral and inhaled PDE4 inhibitors. Bioorg Med Chem Lett 2013; 23:5528-32. [PMID: 24018187 DOI: 10.1016/j.bmcl.2013.08.056] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 08/13/2013] [Indexed: 12/20/2022]
Abstract
The optimization of oxazole-based PDE4 inhibitor 1 has led to the identification of both oral (compound 16) and inhaled (compound 34) PDE4 inhibitors. Selectivity against PDE10/PDE11, off target screening, and in vivo activity in the rat are discussed.
Collapse
Affiliation(s)
- Pauline C Ting
- Department of Chemical Research, Merck Research Laboratories, 126 E. Lincoln Ave., Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Kümmerle AE, Schmitt M, Cardozo SVS, Lugnier C, Villa P, Lopes AB, Romeiro NC, Justiniano H, Martins MA, Fraga CAM, Bourguignon JJ, Barreiro EJ. Design, Synthesis, and Pharmacological Evaluation of N-Acylhydrazones and Novel Conformationally Constrained Compounds as Selective and Potent Orally Active Phosphodiesterase-4 Inhibitors. J Med Chem 2012; 55:7525-45. [DOI: 10.1021/jm300514y] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Arthur E. Kümmerle
- Laboratório de Avaliacão
e Síntese de Substâncias Bioativas (LASSBio), Faculdade
de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 68023, RJ 21944-971, Brazil
- Programa de Pós-Graduação
em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro,
RJ, Brazil
| | - Martine Schmitt
- Laboratoire d’Innovation
Thérapeutique, UMR7200, CNRS, Université de Strasbourg, Faculté de Pharmacie, 74 Route
du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Suzana V. S. Cardozo
- Laboratório de Inflamação,
Departamento de Fisiologia e Farmacodinâmica, Instituto Oswaldo Cruz-Fiocruz, Rio de Janeiro, RJ,
Brazil
- Programa de Pós-Graduação
em Farmacologia e Química Medicinal, Instituto de Ciências
Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Claire Lugnier
- Biophotonique et Pharmacologie,
UMR7213, CNRS, Université de Strasbourg, Faculté de Pharmacie, 74 Route du Rhin, 67400 Illkirch-Graffenstaden,
France
| | - Pascal Villa
- Plate-forme de Chimie Biologie
Integrative
de Strasbourg (PCBIS), UMR3286, CNRS, Université de Strasbourg, Ecole Supérieur de Biotechnologie
et Faculté de Pharmacie, Boulevard Sebastien Brant, 67412 Illkirch-Graffenstaden,
France
| | - Alexandra B. Lopes
- Laboratório de Avaliacão
e Síntese de Substâncias Bioativas (LASSBio), Faculdade
de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 68023, RJ 21944-971, Brazil
- Programa de Pós-Graduação
em Química, Instituto de Química, Universidade Federal do Rio de Janeiro, 21941-902, Rio de Janeiro,
RJ, Brazil
| | - Nelilma C. Romeiro
- Universidade Federal do Rio de Janeiro (UFRJ)-Macaé-Rua Aluísio
da Silva Gomes, 50 Granja dos Cavaleiros, 27930-560 Rio de Janeiro,
RJ, Brazil
| | - Hélène Justiniano
- Biophotonique et Pharmacologie,
UMR7213, CNRS, Université de Strasbourg, Faculté de Pharmacie, 74 Route du Rhin, 67400 Illkirch-Graffenstaden,
France
| | - Marco A. Martins
- Laboratório de Inflamação,
Departamento de Fisiologia e Farmacodinâmica, Instituto Oswaldo Cruz-Fiocruz, Rio de Janeiro, RJ,
Brazil
| | - Carlos A. M. Fraga
- Laboratório de Avaliacão
e Síntese de Substâncias Bioativas (LASSBio), Faculdade
de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 68023, RJ 21944-971, Brazil
- Programa de Pós-Graduação
em Farmacologia e Química Medicinal, Instituto de Ciências
Biomédicas, Universidade Federal do Rio de Janeiro, 21941-902 Rio de Janeiro, RJ, Brazil
| | - Jean-Jacques Bourguignon
- Laboratoire d’Innovation
Thérapeutique, UMR7200, CNRS, Université de Strasbourg, Faculté de Pharmacie, 74 Route
du Rhin, 67400 Illkirch-Graffenstaden, France
| | - Eliezer J. Barreiro
- Laboratório de Avaliacão
e Síntese de Substâncias Bioativas (LASSBio), Faculdade
de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 68023, RJ 21944-971, Brazil
| |
Collapse
|
9
|
Wang L, Yang J, Guo L, Uyeminami D, Dong H, Hammock BD, Pinkerton KE. Use of a soluble epoxide hydrolase inhibitor in smoke-induced chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 2012; 46:614-22. [PMID: 22180869 PMCID: PMC3359909 DOI: 10.1165/rcmb.2011-0359oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 12/12/2011] [Indexed: 11/24/2022] Open
Abstract
Tobacco smoke-induced chronic obstructive pulmonary disease (COPD) is a prolonged inflammatory condition of the lungs characterized by progressive and largely irreversible airflow limitation attributable to a number of pathologic mechanisms, including bronchitis, bronchiolitis, emphysema, mucus plugging, pulmonary hypertension, and small-airway obstruction. Soluble epoxide hydrolase inhibitors (sEHIs) demonstrated anti-inflammatory properties in a rat model after acute exposure to tobacco smoke. We compared the efficacy of sEHI t-TUCB (trans-4-{4-[3-(4-trifluoromethoxy-phenyl)-ureido]-cyclohexyloxy}-benzoic acid) and the phosphodiesterase-4 (PDE4) inhibitor Rolipram (Biomol International, Enzo Life Sciences, Farmingdale, NY) to reduce lung injury and inflammation after subacute exposure to tobacco smoke over a period of 4 weeks. Pulmonary physiology, bronchoalveolar lavage, cytokine production, and histopathology were analyzed to determine the efficacy of sEHI and Rolipram to ameliorate tobacco smoke-induced inflammation and injury in the spontaneously hypertensive rat. Both t-TUCB and Rolipram inhibited neutrophil elevation in bronchoalveolar lavage. sEHI t-TUCB suppressed IFN-γ, while improving lung function by reducing tobacco smoke-induced total respiratory resistance and tissue damping (small-airway and peripheral tissue resistance). Increases in tobacco smoke-induced alveolar airspace size were attenuated by t-TUCB. Rolipram inhibited the production of airway mucus. Both t-TUCB and Rolipram inhibited vascular remodeling-related growth factor. These findings suggest that sEHI t-TUCB has therapeutic potential for treating COPD by improving lung function and attenuating the lung inflammation and emphysematous changes caused by tobacco smoke. To the best of our knowledge, this is the first report to demonstrate that sEHI exerts significant protective effects after repeated, subacute tobacco smoke-induced lung injury in a rat model of COPD.
Collapse
Affiliation(s)
- Lei Wang
- Center for Health and the Environment, and
| | - Jun Yang
- Department of Entomology and Cancer Center, University of California at Davis Medical Center, University of California at Davis, Davis, California
| | - Lei Guo
- Center for Health and the Environment, and
| | | | - Hua Dong
- Department of Entomology and Cancer Center, University of California at Davis Medical Center, University of California at Davis, Davis, California
| | - Bruce D. Hammock
- Department of Entomology and Cancer Center, University of California at Davis Medical Center, University of California at Davis, Davis, California
| | | |
Collapse
|
10
|
Discovery of oxazole-based PDE4 inhibitors with picomolar potency. Bioorg Med Chem Lett 2012; 22:2594-7. [DOI: 10.1016/j.bmcl.2012.01.115] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Revised: 01/26/2012] [Accepted: 01/30/2012] [Indexed: 11/22/2022]
|
11
|
Smith KR, Leonard D, McDonald JD, Tesfaigzi Y. Inflammation, mucous cell metaplasia, and Bcl-2 expression in response to inhaled lipopolysaccharide aerosol and effect of rolipram. Toxicol Appl Pharmacol 2011; 253:253-60. [PMID: 21504754 DOI: 10.1016/j.taap.2011.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 02/16/2011] [Accepted: 04/01/2011] [Indexed: 11/19/2022]
Abstract
Our previous studies have characterized the inflammatory response of intratracheally instilled lipopolysaccharides (LPS) in F344/N rats. To better reflect the environmentally relevant form of LPS exposure, the present study evaluated the inflammatory response of F344/N rats exposed to LPS by inhalation. Rats were exposed by nose-only inhalation to aerosolized LPS at a median particle diameter of 1 μm and a dose range from 0.08 to 480 μg. Animals were euthanized 72 h post exposure and the inflammatory cell counts and differentials, the cytokine/chemokine levels in the bronchoalveolar lavage fluid (BALF), and the changes in intraepithelial stored mucosubstances, mucous cells per mm basal lamina, and Bcl-2-positive mucous cells were quantified. We observed a dose-dependent increase reaching maximum values at the 75 μg LPS dose for the numbers of neutrophils, macrophages and lymphocytes, for the levels of IL-6, IL-1α, IL-1β, TNFα, MCP-1 and GRO-KC. In addition, mucous cell metaplasia and the percentage of Bcl-2-positive mucous cells were increased with an increasing deposited LPS dose. When rats were treated with the phosphodiesterase-4 (PDE4) inhibitor, rolipram (10mg/kg), prior to exposure to aerosolized LPS neutrophil numbers in the BAL were reduced at 8h but not at 24 or 72 h post LPS exposure. These results demonstrate that exposure to aerosolized LPS resulted in a more potent inflammatory response at lower doses and that inflammation was more uniformly distributed throughout the lung compared to inflammation caused by intratracheal LPS instillation. Therefore, this animal model will be useful for screening efficacy of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Kevin R Smith
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM 87108, USA
| | | | | | | |
Collapse
|
12
|
Venkatasamy R, Spina D. Protease inhibitors in respiratory disease: focus on asthma and chronic obstructive pulmonary disease. Expert Rev Clin Immunol 2010; 3:365-81. [PMID: 20477680 DOI: 10.1586/1744666x.3.3.365] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Respiratory diseases, such as asthma and chronic obstructive pulmonary disease (COPD), are a major health burden on society and current treatment modalities for these diseases have not significantly changed over the past 40 years. The only major pharmacological advancement for the treatment of these diseases has been to increase the duration of action of bronchodilators (asthma: salmeterol; COPD: tiotropium bromide) and glucocorticosteroids (asthma: fluticasone propionate) and, increasingly, to formulate these agents in the same delivery device. Despite our increasing understanding of the cell and molecular biology of these diseases, the development of novel treatments remains beyond the reach of the scientific community. Proteases are a family of proteins with diverse biological activity, which are found in abundance within the airways of asthma and COPD, and have been implicated in the pathogenesis of these diseases. The targeting of proteases, including mast cell tryptase, neutrophil elastase and matrix metalloprotease with low-molecular-weight inhibitors, has highlighted the potential role of these enzymes in mediating certain aspects of the disease process in preclinical studies. Several challenges remain regarding the development of protease inhibitors, including the synthesis of highly potent and specific inhibitors, and target validation in man.
Collapse
Affiliation(s)
- Radhakrishnan Venkatasamy
- King's College London, Sackler Institute of Pulmonary Pharmacology, School of Biomedical and Health Science, Pharmaceutical Science Research Division, Guy's Campus, London SE1 1UL, UK.
| | | |
Collapse
|
13
|
Man HW, Schafer P, Wong LM, Patterson RT, Corral LG, Raymon H, Blease K, Leisten J, Shirley MA, Tang Y, Babusis DM, Chen R, Stirling D, Muller GW. Discovery of (S)-N-[2-[1-(3-ethoxy-4-methoxyphenyl)-2-methanesulfonylethyl]-1,3-dioxo-2,3-dihydro-1H-isoindol-4-yl] acetamide (apremilast), a potent and orally active phosphodiesterase 4 and tumor necrosis factor-alpha inhibitor. J Med Chem 2009; 52:1522-4. [PMID: 19256507 DOI: 10.1021/jm900210d] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In this communication, we report the discovery of 1S (apremilast), a novel potent and orally active phosphodiesterase 4 (PDE4) and tumor necrosis factor-alpha inhibitor. The optimization of previously reported 3-(1,3-dioxo-1,3-dihydroisoindol-2-yl)-3-(3,4-dimethoxyphenyl)propionic acid PDE4 inhibitors led to this series of sulfone analogues. Evaluation of the structure-activity relationship of substitutions on the phthalimide group led to the discovery of an acetylamino analogue 1S, which is currently in clinical trials.
Collapse
Affiliation(s)
- Hon-Wah Man
- Drug Discovery Department, Celgene Corporation, 86 Morris Avenue, Summit, New Jersey 07901, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Sharma R, Kaundal RK, Sharma SS. Amelioration of pulmonary dysfunction and neutrophilic inflammation by PPAR gamma agonist in LPS-exposed guinea pigs. Pulm Pharmacol Ther 2008; 22:183-9. [PMID: 19073273 DOI: 10.1016/j.pupt.2008.11.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 09/28/2008] [Accepted: 11/24/2008] [Indexed: 12/16/2022]
Abstract
Airway dysfunction and pulmonary neutrophilic inflammation are the major characteristics of inflammatory conditions of lungs like chronic obstructive pulmonary disease (COPD). Lipopolysaccharide (LPS), a constituent of cigarette smoke, has been identified as the most important risk factor for COPD development. Inhalation exposure to LPS or cigarette smoke elicits an inflammatory response accompanied by airway hyperresponsiveness, elevated proinflammatory mediators and inflammatory cells similar to COPD. In the present study, we have evaluated the effects of pioglitazone, a peroxisome proliferator-activated receptor gamma (PPAR gamma) agonist, in LPS-induced pulmonary dysfunction, inflammatory changes and oxidative stress in guinea pigs. Inhalation exposure to nebulised LPS (30 microg ml(-1)) resulted in significant increase in the breathing frequency and bronchoconstriction accompanied with a significant decrease in tidal volume. Our results demonstrated that the LPS-induced pulmonary dysfunction was temporally associated with neutrophil infiltration as evident from heavy neutrophilia, increased TNFalpha in bronchoalveolar lavage fluid (BAL), elevated myeloperoxidase (MPO) level and histology of the lung tissue. Exposure to LPS also produced significant increase in tissue malondialdehyde (MDA) level indicating underlying oxidative stress. The results also reveal that pioglitazone (3, 10 and 30 mg kg(-1), p.o.) is effective in abrogating the pulmonary dysfunction by attenuating neutrophilia, TNFalpha release and oxidative stress in LPS-induced model of acute lung inflammation. Results from the present study have added to the emergent body of evidence that PPAR gamma agonists are effective in the therapy of inflammatory disease of the lungs.
Collapse
Affiliation(s)
- Rohini Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, S.A.S. Nagar (Mohali), Punjab 160062, India
| | | | | |
Collapse
|
15
|
Michel O, Dentener M, Cataldo D, Cantinieaux B, Vertongen F, Delvaux C, Murdoch RD. Evaluation of oral corticosteroids and phosphodiesterase-4 inhibitor on the acute inflammation induced by inhaled lipopolysaccharide in human. Pulm Pharmacol Ther 2007; 20:676-83. [PMID: 17045826 DOI: 10.1016/j.pupt.2006.08.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 06/22/2006] [Accepted: 08/22/2006] [Indexed: 11/18/2022]
Abstract
BACKGROUND Endotoxins are pro-inflammatory substances present in the environment. In man, inhalation of its purified derivative lipopolysaccharide (LPS) induces inflammation related to macrophages and neutrophils. Corticosteroids and phosphodiesterase (PDE)-4 inhibitors have inhibiting effects on macrophages and neutrophils, respectively. This study investigated the effect of prednisolone and of the PDE-4 inhibitor cilomilast on the LPS-induced acute inflammation. METHODS The study was a placebo-controlled, double-blind crossover design. On three occasions, at 2 weeks interval, 16 healthy subjects inhaled 50 microg LPS after a 6-day treatment with cilomilast (15 mg bd), prednisolone (10 mg bd) or placebo. For the assessment of the inflammatory response, induced sputum was obtained before inclusion and 6h post-LPS while blood samples were collected before, 6 and 24 h post-LPS. RESULTS Inhaled LPS induced an increase in sputum neutrophils (p<0.0001), logMMP-9 (p<0.05), logMMP-9/TIMP-1 (p<0.01) and logTNF-alpha (p<0.02). At the blood level there were significant rise in neutrophilia (p<0.001), E-selectin (p<0.02), C-reactive protein (CRP) (p<0.001) and LPS-binding protein (p<0.001). There was both a slight, but not significant, increase in body temperature and decrease in forced expiratory volume in 1 s (FEV(1)). Neither prednisolone nor cilomilast had protective effect on the LPS-induced airways' inflammation. The LPS-induced CRP acute-phase protein of inflammation (0.58+/-0.13 and 3.52+/-0.41 mg/dL, before and after LPS, respectively) was significantly inhibited by a pre-treatment with prednisolone (1.39+/-0.32 mg/dL, p<0.01) and attenuated (2.65+/-0.30 mg/dL, p=0.09) with cilomilast. CONCLUSION In healthy subjects, while the LPS-induced airways' inflammation was not modified either by oral prednisolone or by PDE-4 inhibitor cilomilast (at actual dosage), the LPS-induced acute phase of blood inflammation was reduced by prednisolone.
Collapse
Affiliation(s)
- Olivier Michel
- Clinics of Allergology and Respiratory Diseases, CHU Saint-Pierre (ULB-CP404), Rue Haute 322, B-1000 Brussels, Belgium.
| | | | | | | | | | | | | |
Collapse
|
16
|
Kuang R, Shue HJ, Blythin DJ, Shih NY, Gu D, Chen X, Schwerdt J, Lin L, Ting PC, Zhu X, Aslanian R, Piwinski JJ, Xiao L, Prelusky D, Wu P, Zhang J, Zhang X, Celly CS, Minnicozzi M, Billah M, Wang P. Discovery of a highly potent series of oxazole-based phosphodiesterase 4 inhibitors. Bioorg Med Chem Lett 2007; 17:5150-4. [PMID: 17683932 DOI: 10.1016/j.bmcl.2007.06.092] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2007] [Revised: 06/28/2007] [Accepted: 06/29/2007] [Indexed: 10/23/2022]
Abstract
Substituted quinolyl oxazoles were discovered as a novel and highly potent series of phosphodiesterase 4 (PDE4) inhibitors. Structure-activity relationship studies revealed that the oxazole core, with 4-carboxamide and 5-aminomethyl groups, is a novel PDE4 inhibitory pharmacophore. Selectivity profiles and in vivo biological activity are also reported.
Collapse
Affiliation(s)
- Rongze Kuang
- Department of Chemical Research, Schering-Plough Research Institute, 2015 Galloping Hill Road, Kenilworth, NJ 07033, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Chapman RW, Minnicozzi M, Celly CS, Phillips JE, Kung TT, Hipkin RW, Fan X, Rindgen D, Deno G, Bond R, Gonsiorek W, Billah MM, Fine JS, Hey JA. A Novel, Orally Active CXCR1/2 Receptor Antagonist, Sch527123, Inhibits Neutrophil Recruitment, Mucus Production, and Goblet Cell Hyperplasia in Animal Models of Pulmonary Inflammation. J Pharmacol Exp Ther 2007; 322:486-93. [PMID: 17496165 DOI: 10.1124/jpet.106.119040] [Citation(s) in RCA: 106] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Sch527123 [2-hydroxy-N,N-dimethyl-3-[[2-[[1(R)-(5-methyl-2-furanyl)propyl]amino]-3,4-dioxo-1-cyclobuten-1-yl]amino]ben-zamide] is a potent, selective antagonist of the human CXCR1 and CXCR2 receptors (Gonsiorek et al., 2007). Here we describe its pharmacologic properties at rodent CXCR2 and at the CXCR1 and CXCR2 receptors in the cynomolgus monkey, as well as its in vivo activity in models demonstrating prominent pulmonary neutrophilia, goblet cell hyperplasia, and mucus production. Sch527123 bound with high affinity to the CXCR2 receptors of mouse (K(d) = 0.20 nM), rat (K(d) = 0.20 nM), and cynomolgus monkey (K(d) = 0.08 nM) and was a potent antagonist of CXCR2-mediated chemotaxis (IC(50) approximately 3-6 nM). In contrast, Sch527123 bound to cynomolgus CXCR1 with lesser affinity (K(d) = 41 nM) and weakly inhibited cynomolgus CXCR1-mediated chemotaxis (IC(50) approximately 1000 nM). Oral treatment with Sch527123 blocked pulmonary neutrophilia (ED(50) = 1.2 mg/kg) and goblet cell hyperplasia (32-38% inhibition at 1-3 mg/kg) in mice following the intranasal lipopolysaccharide (LPS) administration. In rats, Sch527123 suppressed the pulmonary neutrophilia (ED(50) = 1.8 mg/kg) and increase in bronchoalveolar lavage (BAL) mucin content (ED(50) =<0.1 mg/kg) induced by intratracheal (i.t.) LPS. Sch527123 also suppressed the pulmonary neutrophilia (ED(50) = 1.3 mg/kg), goblet cell hyperplasia (ED(50) = 0.7 mg/kg), and increase in BAL mucin content (ED(50) = <1 mg/kg) in rats after i.t. administration of vanadium pentoxide. In cynomolgus monkeys, Sch527123 reduced the pulmonary neutrophilia induced by repeat bronchoscopy and lavage (ED(50) = 0.3 mg/kg). Therefore, Sch527123 may offer benefit for the treatment of inflammatory lung disorders in which pulmonary neutrophilia and mucus hypersecretion are important components of the underlying disease pathology.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/metabolism
- Anti-Inflammatory Agents/pharmacokinetics
- Anti-Inflammatory Agents/therapeutic use
- Benzamides/metabolism
- Benzamides/pharmacology
- Benzamides/therapeutic use
- Biological Availability
- Bronchitis/chemically induced
- Bronchitis/drug therapy
- Bronchitis/metabolism
- Bronchoalveolar Lavage
- Bronchoalveolar Lavage Fluid/chemistry
- Bronchoalveolar Lavage Fluid/cytology
- Bronchoscopy
- Cell Line
- Cell Membrane/metabolism
- Chemokines, CXC/analysis
- Chemokines, CXC/metabolism
- Chemotaxis/drug effects
- Chemotaxis, Leukocyte/drug effects
- Cyclobutanes/metabolism
- Cyclobutanes/pharmacology
- Cyclobutanes/therapeutic use
- Disease Models, Animal
- Goblet Cells/pathology
- Hyperplasia/drug therapy
- Hyperplasia/pathology
- Lipopolysaccharides/pharmacology
- Lung/metabolism
- Lung/pathology
- Macaca fascicularis
- Male
- Mice
- Mice, Inbred BALB C
- Mucins/analysis
- Mucins/metabolism
- Mucus/metabolism
- Neutrophils/pathology
- Rats
- Rats, Sprague-Dawley
- Receptors, Interleukin-8A/antagonists & inhibitors
- Receptors, Interleukin-8A/genetics
- Receptors, Interleukin-8A/metabolism
- Receptors, Interleukin-8B/antagonists & inhibitors
- Receptors, Interleukin-8B/genetics
- Receptors, Interleukin-8B/metabolism
- Vanadium Compounds/pharmacology
Collapse
Affiliation(s)
- Richard W Chapman
- Schering Plough Research Institute, Pulmonary and Peripheral Neurobiology, Kenilworth, NJ 07033, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Siaud P, Maurel D, Lucciano M, Kosa E, Cazals Y. Enhanced cochlear acoustic sensitivity and susceptibility to endotoxin are induced by adrenalectomy and reversed by corticosterone supplementation in rat. Eur J Neurosci 2007; 24:3365-71. [PMID: 17229085 DOI: 10.1111/j.1460-9568.2006.05224.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glucocorticoid receptors are widely distributed in the cochlea but their role remains poorly known. Previous studies provided contradictory reports on a possible cochlear acoustic hypersensitivity induced by adrenal insufficiency, while several experiments agree on a significant action of glucocorticoid receptors in adverse conditions such as acoustic trauma and restraint stress. The present experiments confirmed a cochlear acoustic hypersensitivity induced by adrenalectomy and reversed by corticosterone supplementation. These observations point to a significant role of corticosteroids in basal cochlear functioning. The glucocorticoids are known to be essential for limiting and resolving inflammatory processes. The endotoxin Escherichia coli lipopolysaccharide is widely used to induce inflammatory reactions. However, in various organs several toxic processes of this endotoxin are not influenced by glucocorticoids. From previous experiments on the cochlea there is no evidence that glucocorticoids are an essential factor against endotoxin cochlear toxicity. In the present experiments it was found that adrenalectomy greatly increased the cochlear susceptibility to endotoxin; the effect was reversed by providing corticosterone supplementation. This shows the essential role of corticosterone in this cochlear inflammation model. In previous studies local administration (at the cochlear base) of endotoxin was used and losses of cochlear acoustic sensitivity were found predominantly at high frequencies; in contrast, the systemic injection used in this study produced a cochlear loss of acoustic sensitivity at all frequencies, indicating a uniform cochlear sensitivity to the toxic effects of endotoxin.
Collapse
Affiliation(s)
- Philippe Siaud
- Inserm EPI9902, Faculté de Médecine Marseille Nord, Boulevard P Dramard 13916 Marseille Cedex 20, France
| | | | | | | | | |
Collapse
|
19
|
Nénan S, Lagente V, Planquois JM, Hitier S, Berna P, Bertrand CP, Boichot E. Metalloelastase (MMP-12) induced inflammatory response in mice airways: Effects of dexamethasone, rolipram and marimastat. Eur J Pharmacol 2007; 559:75-81. [PMID: 17234180 DOI: 10.1016/j.ejphar.2006.11.070] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Revised: 11/29/2006] [Accepted: 11/30/2006] [Indexed: 11/18/2022]
Abstract
Direct instillation of a recombinant human form of MMP-12 (rhMMP-12) in mice airways elicited an early inflammatory response characterized by neutrophil influx, cytokine release and gelatinase activation followed by a delayed response, mainly characterized by macrophage recruitment. As this experimental model of lung inflammation partially mimics some features of chronic obstructive pulmonary disease (COPD), we have investigated the effects of treatment by anti-inflammatory compounds, dexamethasone and rolipram and a non-specific matrix metalloproteinase (MMP) inhibitor, marimastat. The compounds were administrated orally, 1 h before rhMMP-12 instillation (8 x 10(-3) U/mouse). Total and differential cell counts were evaluated in the bronchoalveolar lavage fluids. Cytokines and MMP-9 were quantified in bronchoalveolar lavage fluids and in lung homogenate supernatants. Marimastat (100 mg/kg), dexamethasone (10 mg/kg) and rolipram (0.1 and 0.3 mg/kg) were able to decrease significantly neutrophil recruitment at 4 and 24 h after rhMMP-12 instillation, but only marimastat (30 and 100 mg/kg) was effective at decreasing the macrophage recruitment occurring at day 7. Marimastat (100 mg/kg), dexamethasone (10 mg/kg) and rolipram (0.3 mg/kg) reduced significantly IL-6, KC/CXCL1, MIP-1alpha/CCL3 and MMP-9 levels in bronchoalveolar lavage fluid. Similar results were obtained in lung homogenates except with rolipram. Dexamethasone and rolipram were able to inhibit the early inflammatory response but were ineffective to limit the macrophage influx. In contrast, marimastat was able to reduce early and late response. These data indicate that MMP-12 instillation in mice could highlight some of the inflammatory response seen in COPD and could be used for the pharmacological evaluation of new anti-inflammatory mechanisms of action.
Collapse
Affiliation(s)
- Soazig Nénan
- INSERM U620, University of Rennes 1, Rennes, France
| | | | | | | | | | | | | |
Collapse
|
20
|
Koike T, Nadeen Qutab M, Tsuchida M, Takekubo M, Saito M, Hayashi JI. Pretreatment with olprinone hydrochloride, a phosphodiesterase III inhibitor, attenuates lipopolysaccharide-induced lung injury via an anti-inflammatory effect. Pulm Pharmacol Ther 2007; 21:166-71. [PMID: 17434327 DOI: 10.1016/j.pupt.2007.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2006] [Revised: 12/26/2006] [Accepted: 01/20/2007] [Indexed: 10/23/2022]
Abstract
PURPOSE Acute respiratory distress syndrome is characterized by neutrophil accumulation in the lungs and the activation of several cytokines produced by macrophages. Olprinone hydrochloride, a specific phosphodiesterase III inhibitor, has anti-inflammatory effects and inhibits the activation of macrophages, in addition to its inotropic and vasodilatory effects. The purpose of this study was to examine the beneficial effects of olprinone on lipopolysaccharide (LPS)-induced pulmonary inflammation. MATERIALS AND METHODS Lung inflammation was produced by intravenous LPS injection into rats. The rats were divided into four groups: a vehicle group in which normal saline was injected, an olprinone group in which olprinone was injected at a dose of 0.2mg/kg, a dexamethasone group in which dexamethasone was injected at a dose of 5mg/kg, and a control group. In each group, drug was injected intraperitoneally 30 min before the intravenous administration of LPS. The blood was obtained at 1h and then animals were sacrificed at 6h and blood and lung specimen were obtained for cytokine analysis and pathological examination. On another set of experiment, bronchioloalveolar lavage (BAL) was performed for cytokine analysis of BAL fluid. The macrophages isolated from normal rat by BAL were cultured in vitro with the presence of LPS and olprinone or dexamethasone, and supernatant was collected. The levels of several cytokines in the serum, in the BAL fluid, and in the culture supernatant were determined. RESULTS The animals injected with LPS were found to have an influx of neutrophils in the lungs, and inflammatory cytokines, such as TNF-alpha and IL-6, and anti-inflammatory cytokine IL-10 were produced. Pretreatment with olprinone or dexamethasone significantly inhibited the LPS-induced neutrophil influx into the lungs, suppressed inflammatory cytokines TNF-alpha and IL-6. The level of anti-inflammatory cytokine IL-10 increased in an olprinone group. The inhibition of TNF-alpha and IL-6, and the augmentation of IL-10 release were also observed in in vitro culture of isolated rat alveolar macrophages when olprinone (10(-5)mol/ml) and LPS (10 microg/ml) were cultured together. However, the level of IL-10 in serum and culture supernatant was suppressed in a dexamesathone group. CONCLUSION LPS-induced lung inflammation is strongly inhibited by olprinone accompanying the enhancement of IL-10 and the inhibition of inflammatory cytokines. Results of the in vitro experiment suggest that alveolar macrophages may play an important role in ameliorating LPS-induced lung inflammation and the mechanism of its effect is different from that of steroid.
Collapse
Affiliation(s)
- Terumoto Koike
- Division of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Niigata 951-8510, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Wollin L, Bundschuh DS, Wohlsen A, Marx D, Beume R. Inhibition of airway hyperresponsiveness and pulmonary inflammation by roflumilast and other PDE4 inhibitors. Pulm Pharmacol Ther 2006; 19:343-52. [PMID: 16257550 DOI: 10.1016/j.pupt.2005.09.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2005] [Revised: 09/15/2005] [Accepted: 09/19/2005] [Indexed: 10/25/2022]
Abstract
Roflumilast is an oral, once-daily phosphodiesterase 4 (PDE4) inhibitor with anti-inflammatory activity. We compared the anti-inflammatory effects of roflumilast with those of PDE4 inhibitors rolipram, piclamilast, and cilomilast in ovalbumin (OVA)-sensitized and challenged Brown-Norway rats. Animals were treated orally 1h before OVA challenge with roflumilast (0.3, 1.0, and 3.0mg/kg), rolipram (0.8, 2.8, and 8.3mg/kg), piclamilast (10.0, 20.0, and 30.0mg/kg), or cilomilast (10.3, 34.3, and 103.0mg/kg). Airway hyperresponsiveness (AHR) against adenosine was investigated by measuring airway resistance 200min after OVA challenge. Subsequently, neutrophil influx and tumor necrosis factor-alpha (TNF-alpha) release in the lungs were determined by bronchoalveolar lavage. Direct bronchodilation at the time point of AHR assessment by PDE4 inhibitors was examined in serotonin-challenged animals. Evaluation of neutropenic animals or treatment with anti-TNF-alpha antibody revealed that AHR was independent of neutrophil accumulation or TNF-alpha release. Roflumilast (50% inhibitory dose [ID(50)]=1.5mg/kg) inhibited AHR 3-, 16-, and 27-fold more potently than rolipram, piclamilast, and cilomilast, respectively. Likewise, roflumilast was a more potent inhibitor of neutrophil influx (ID(50)=0.9mg/kg) than rolipram (ID(50)=6.9mg/kg), piclamilast (ID(50)=28.1mg/kg), or cilomilast (ID(50)=37.7mg/kg). Roflumilast, rolipram, and piclamilast-but not cilomilast-suppressed OVA-induced TNF-alpha release in a dose-dependent manner. Roflumilast (ID(50)=0.9mg/kg) exhibited 9- and 23-fold more potent inhibition of TNF-alpha release than rolipram and piclamilast, respectively. Roflumilast did not inhibit serotonin-induced bronchoconstriction 4.5h after administration, suggesting that inhibition of AHR by roflumilast results from anti-inflammatory, not bronchodilatory, effects. This study suggests that roflumilast has anti-inflammatory action and provides rationale for the investigation of roflumilast in asthmatic patients.
Collapse
Affiliation(s)
- L Wollin
- ALTANA Pharma, Byk-Gulden-Strasse 2, 78467 Konstanz, Germany.
| | | | | | | | | |
Collapse
|
22
|
McCluskie K, Klein U, Linnevers C, Ji YH, Yang A, Husfeld C, Thomas GR. Phosphodiesterase Type 4 Inhibitors Cause Proinflammatory Effects in Vivo. J Pharmacol Exp Ther 2006; 319:468-76. [PMID: 16861399 DOI: 10.1124/jpet.106.105080] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Phosphodiesterase type 4 (PDE(4)) inhibitors are currently being evaluated as potential therapies for inflammatory airway diseases. However, this class of compounds has been shown to cause an arteritis/vasculitis of unknown etiology in rats and cynomolgus monkeys. Studies in rodents have demonstrated the anti-inflammatory effects of PDE(4) inhibitors on lipopolysaccharide (LPS)-induced airway inflammation. The aim of this work was to assess the direct effects of PDE(4) inhibitors on inflammatory cells and cytokine levels in the lung in relation to therapeutic effects. The effects of the PDE(4) inhibitors 3-cyclo-propylmethoxy-4-difluoromethoxy-N-[3,5-di-chloropyrid-4-yl]-benzamide (roflumilast) and 3-(cyclopentyloxy)-N-(3,5-dichloro-4-pyridyl)-4-methoxybenzamide (piclamilast) were assessed in vivo, using BALB/c mice, and in vitro, in unstimulated human endothelial and epithelial cell lines. In BALB/c mice, LPS challenge caused an increase in neutrophils in bronchoalveolar lavage (BAL) and lung tissue and BAL tumor necrosis factor-alpha levels, which were inhibited by treatment with either roflumilast or piclamilast (30-100 mg/kg subcutaneously). However, roflumilast and piclamilast alone (100 mg/kg) caused a significant increase in plasma and lung tissue keratinocyte-derived chemokine (KC) levels, and lung tissue neutrophils. In vitro, both piclamilast and roflumilast caused an increase in interleukin (IL)-8 release from human umbilical vein endothelial cells but not BEAS-2B cells, suggesting that one source of the increased KC may be endothelial cells. At doses that antagonized an LPS-induced inflammatory response, the PDE(4) inhibitors possessed proinflammatory activities in the lung that may limit their therapeutic potential. The proinflammatory cytokines KC and IL-8 therefore may provide surrogate biomarkers, both in preclinical animal models and in the clinic, to assess potential proinflammatory effects of this class of compounds.
Collapse
Affiliation(s)
- Kerryn McCluskie
- Department of Pharmacology, Theravance Inc., South San Francisco, California, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Chronic obstructive pulmonary disease (COPD) is a multicomponent, chronic inflammatory disease of the lungs with systemic complications. The majority of the inflammation occurs in the peripheral airways and lung parenchyma. It is a progressive disease, leading to disability and eventual death, despite conventional therapy. Inflammatory activity can be reduced by increasing intracellular cyclic adenosine-3',5'-monophosphate (cAMP) through inhibition of phosphodiesterase (PDE) IV, the principal PDE isoenzyme within pro-inflammatory cells, including eosinophils, mast cells, macrophages, lymphocytes, neutrophils and epithelial cells. PDE IV inhibition also has other effects, including relaxation of airway smooth muscle, suppression of smooth muscle mitogenesis and modulation of excitatory activity in pulmonary nerves. Cilomilast is a systemically available, second-generation, selective PDE IV inhibitor. It retains the therapeutic activity of the first-generation PDE IV inhibitors but lacks their profound emetic effect. Cilomilast is the first drug to demonstrate a reduction of tissue cells considered central to the ongoing inflammatory process (macrophages and CD8+ lymphocytes) in patients with stable COPD. Cilomilast is completely absorbed following oral administration and has negligible first-pass metabolism. It exhibits linear pharmacokinetics, with low between-subject variability. Cilomilast is highly protein bound (99.4%), but this binding is concentration-independent at clinically relevant doses, and it has a small volume of distribution at steady state (17L). Plasma clearance (approximately 2 L/h) is almost entirely metabolic, through multiple parallel pathways. Its terminal elimination half-life is approximately 6.5 hours and steady state is rapidly achieved with twice-daily administration. The most abundant metabolite, formed by the action of cytochrome P450 2C8, has <10% of the activity of the parent molecule. Cilomilast pharmacokinetics in COPD patients were consistent with those in healthy subjects. Smoking, age and ethnicity had no clinically relevant effects. Total plasma cilomilast pharmacokinetic parameters did not change significantly with renal or hepatic impairment, but concentrations of unbound cilomilast increased with declining renal or hepatic function. Cilomilast had no clinically relevant interactions with a range of drugs likely to be coadministered to patients with COPD, with the exception of erythromycin where concurrent administration with cilomilast was associated with an increased incidence of gastrointestinal adverse events, a pharmacodynamic interaction predicted by their secondary pharmacology. Nausea was the principal adverse reaction seen in healthy subjects taking cilomilast, but this was reduced by administration with food or by use of simple dose-escalation regimens. Cilomilast has not shown a propensity for any of the serious cardiac or neurological adverse effects associated with theophylline. Cilomilast exhibits favourable and predictable pharmacokinetics, has few clinically relevant drug-drug interactions and has demonstrated effects on measures of inflammation of potential benefit in the treatment of COPD. It is generally well tolerated and has not generated safety concerns in any clinical study.
Collapse
Affiliation(s)
- Geoff Down
- Respiratory Clinical Pharmacology, GlaxoSmithKline Research and Development, Greenford, England.
| | | | | | | |
Collapse
|
24
|
Lavoie JP, Pasloske K, Joubert P, Cordeau ME, Mancini J, Girard Y, Friesen RW, Frenette R, Blouin M, Young RN, Hickey G. Lack of Clinical Efficacy of a Phosphodiesterase-4 Inhibitor for Treatment of Heaves in Horses. J Vet Intern Med 2006. [DOI: 10.1111/j.1939-1676.2006.tb02838.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
25
|
Abstract
PURPOSE OF REVIEW Chronic obstructive pulmonary disease (COPD) is a disease state characterized by airflow limitation that is usually progressive. In addition, an abnormal inflammatory response of the lungs to noxious particles or gases can be seen throughout the airways, parenchyma, and pulmonary vasculature. So far, anti-inflammatory medications (eg, inhaled corticosteroids) have failed to show a major effect on the decline of lung function in COPD patients. Novel anti-inflammatory therapies such as selective phosphodiesterase 4 (PDE4) inhibitors are in clinical development. Their potential role in the management of COPD is described in this review. RECENT FINDINGS Some of the selective PDE4 inhibitors have demonstrated in vitro and in vivo anti-inflammatory activity on cells commonly linked to airway inflammation in COPD, such as neutrophils. While these agents seem to offer only a modest improvement in lung function compared with other bronchodilators, their anti-inflammatory effects appear to provide some substantial benefits in reducing exacerbations and improving health-related quality of life. SUMMARY Based on the available data, the second generation of selective PDE4 inhibitors will likely provide additional therapeutic options for the management of COPD. These agents may become an important tool in the treatment of this disease, since they target three important components of COPD: airway obstruction, inflammation, and structural changes.
Collapse
Affiliation(s)
- Francisco J Soto
- Division of Pulmonary and Critical Care Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA.
| | | |
Collapse
|
26
|
Sanz MJ, Nabah YNA, Cerdá-Nicolás M, O'Connor JE, Issekutz AC, Cortijo J, Morcillo EJ. Erythromycin exerts in vivo anti-inflammatory activity downregulating cell adhesion molecule expression. Br J Pharmacol 2005; 144:190-201. [PMID: 15665859 PMCID: PMC1575992 DOI: 10.1038/sj.bjp.0706021] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. Macrolides have long been used as anti-bacterial agents; however, there is some evidence that may exert anti-inflammatory activity. Therefore, erythromycin was used to characterize the mechanisms involved in their in vivo anti-inflammatory activity. 2. Erythromycin pretreatment (30 mg kg(-1) day(-1) for 1 week) reduced the lipopolysaccharide (LPS; intratracheal, 0.4 mg kg(-1))-induced increase in neutrophil count and elastase activity in the bronchoalveolar lavage fluid (BALF) and lung tissue myeloperoxidase activity, but failed to decrease tumor necrosis factor-alpha and macrophage-inflammatory protein-2 augmented levels in BALF. Erythromycin pretreatment also prevented lung P-selectin, E-selectin, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) mRNA upregulation in response to airway challenge with LPS. 3. Mesentery superfusion with LPS (1 mug ml(-1)) induced a significant increase in leukocyte-endothelial cell interactions at 60 min. Erythromycin pretreatment abolished the increases in these parameters. 4. LPS exposure of the mesentery for 4 h caused a significant increase in leukocyte rolling flux, adhesion and emigration, which were inhibited by erythromycin by 100, 93 and 95%, respectively. 5. Immunohistochemical analysis showed that LPS exposure of the mesentery for 4 h caused a significant enhancement in P-selectin, E-selectin, ICAM-1 and VCAM-1 expression that was downregulated by erythromycin pretreatment. 6. Flow cytometry analysis indicated that erythromycin pretreatment inhibited LPS-induced CD11b augmented expression in rat neutrophils. 7. In conclusion, erythromycin inhibits leukocyte recruitment in the lung and this effect appears mediated through downregulation of CAM expression. Therefore, macrolides may be useful in the control of neutrophilic pulmonary diseases.
Collapse
Affiliation(s)
- María-Jesús Sanz
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Avda. Blasco Ibañez 15, Valencia 46010, Spain
| | - Yafa Naim Abu Nabah
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Avda. Blasco Ibañez 15, Valencia 46010, Spain
| | | | | | - Andrew C Issekutz
- Departments of Pediatrics, Pathology, Microbiology and Immunology, Dalhousie University, Halifax, Canada
| | - Julio Cortijo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Avda. Blasco Ibañez 15, Valencia 46010, Spain
- Valencia General Hospital Research Foundation, Valencia, Spain
| | - Esteban J Morcillo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Avda. Blasco Ibañez 15, Valencia 46010, Spain
- Author for correspondence:
| |
Collapse
|
27
|
Phillips JE, Case NR, Celly C, Chapman RW, Hey JA, Minnicozzi M. An enzyme-linked immunosorbent assay (ELISA) for the determination of mucin levels in bronchoalveolar lavage fluid. J Pharmacol Toxicol Methods 2005; 53:160-7. [PMID: 16507438 DOI: 10.1016/j.vascn.2005.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2004] [Accepted: 03/01/2005] [Indexed: 10/25/2022]
Abstract
INTRODUCTION A method to measure the mucin concentration in bronchoalveolar lavage (BAL) fluid was developed to aid efforts to identify pharmacologically the mechanisms that modulate pathophysiological mucin secretion. Mucins are the major macromolecular components of mucus. In the airways, mucus is the first line of defense against inhaled microorganisms (infection) and particulates (irritation). METHODS An enzyme-linked immunosorbent assay (ELISA) was developed, comparing two monoclonal anti-mucin antibodies (A10G5 and 45M1) raised to human mucin, to quantify the mucin in BAL fluid from animal models of pulmonary inflammation. To validate the ELISA method, rats were exposed to ovalbumin (OVA, in sensitized rats), lipopolysaccharide (LPS), vanadium pentoxide (V(2)O(5)), or saline. One hundred microliters of BAL fluid was analyzed for mucin concentration. Pooled BAL fluid from untreated rats was used as an internal "plate standard", as a standard mucin that cross-reacts with A10G5 was unavailable. RESULTS We found both antibodies reacted with rat, human, and guinea-pig mucin; where the 45M1 antibody also reacted with the mucin in porcine BAL, while A10G5 did not. We determined the mucin concentration in each BAL fluid sample relative to the standard, defined as a mucin concentration of 100 plate units. BAL fluid from LPS (218+/-25 plate units, n=5), OVA (386+/-31, n=3), V(2)O(5) (1208+/-450, n=6) challenged rats displayed significantly elevated mucin concentration over their saline controls (126+/-22, n=12). Subsequently, the 45M1 antibody displayed immunoreactivity with a commercially available crude preparation of porcine stomach mucin, allowing us to calculate the concentration of mucin directly compared to the known concentration of the porcine stomach mucin standard. Both the 45M1 and A10G5 based ELISA assays detected higher mucin content in the saline challenged rat than the saline challenged guinea pig BAL. DISCUSSION The recent availability of the 45M1 antibody and the use of the crude purification of porcine stomach mucin as a reference standard should allow for direct comparison of mucin concentration in BAL (and other fluids).
Collapse
Affiliation(s)
- Jonathan E Phillips
- Pulmonary and Peripheral Neurobiology, Schering-Plough Research Institute, 2015 Galloping Hill Road (M/S 1700), Kenilworth, NJ 07033, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Sanz MJ, Cortijo J, Morcillo EJ. PDE4 inhibitors as new anti-inflammatory drugs: effects on cell trafficking and cell adhesion molecules expression. Pharmacol Ther 2005; 106:269-97. [PMID: 15922015 DOI: 10.1016/j.pharmthera.2004.12.001] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2004] [Indexed: 01/15/2023]
Abstract
Phosphodiesterase 4 (PDE4) is a major cyclic AMP-hydrolyzing enzyme in inflammatory and immunomodulatory cells. The wide range of inflammatory mechanisms under control by PDE4 points to this isoenzyme as an attractive target for new anti-inflammatory drugs. Selective inhibitors of PDE4 have demonstrated a broad spectrum of anti-inflammatory activities including the inhibition of cellular trafficking and microvascular leakage, cytokine and chemokine release from inflammatory cells, reactive oxygen species production, and cell adhesion molecule expression in a variety of in vitro and in vivo experimental models. The initially detected side effects, mainly nausea and emesis, appear at least partially overcome by the 'second generation' PDE4 inhibitors, some of which like roflumilast and cilomilast are in the later stages of clinical development for treatment of chronic obstructive pulmonary disease. These new drugs may also offer opportunities for treatment of other inflammatory diseases.
Collapse
Affiliation(s)
- María Jesús Sanz
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Avenida Blasco Ibáñez 15, E-46010 Valencia, Spain
| | | | | |
Collapse
|
29
|
Spond J, Billah MM, Chapman RW, Egan RW, Hey JA, House A, Kreutner W, Minnicozzi M. The role of neutrophils in LPS-induced changes in pulmonary function in conscious rats. Pulm Pharmacol Ther 2004; 17:133-40. [PMID: 15123222 DOI: 10.1016/j.pupt.2004.01.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2003] [Revised: 12/05/2003] [Accepted: 01/07/2004] [Indexed: 10/26/2022]
Abstract
We have previously reported on a model of lipopolysaccharide (LPS)-induced pulmonary inflammation in rats, where LPS-challenged animals develop a significant pulmonary neutrophilia and mucus hypersecretion. In the current studies, we utilized whole body plethysmography and computer assisted data acquisition to examine changes in pulmonary parameters, e.g. frequency (f) tidal volume and Penh as a measure of bronchoconstriction, due to LPS-challenge in conscious rats. Compared to saline challenge, LPS-challenged rats displayed a significant increase in (f) which began within 30 min, peaked by 2 h and remained elevated up to 24 h. Mirroring this increase in (f) was a decrease in the observed tidal volume of LPS-challenged rats. Additionally, compared to saline challenge, LPS-challenge provoked a significant and spontaneous bronchoconstriction, as measured by Penh, 2 h after challenge. In order to further understand these observed LPS-induced pulmonary changes, we utilized two classes of pulmonary obstructive disease standards, namely, bronchodilators and anti-inflammatory agents, and examined their ability to affect the spontaneous bronchoconstriction and the increase in (f) seen at two discrete time points, i.e. 2 and 24 h after LPS-challenge. While ineffective on either the 2 h increase in (f) or the LPS-induced inflammation, animals pretreated with salbutamol (10 mg/kg, p.o.) were protected from the increase in (f) seen at the 24 h time point after LPS-challenge. In contrast, when animals were pretreated with theophylline (10 mg/kg, p.o.) no effect on the LPS-induced pulmonary inflammation or increase in (f) was noted. Meanwhile, in animals pretreated with either betamethasone (3 mg/kg, p.o.) or SB207499 (10 mg/kg, p.o.), a PDE4 inhibitor, doses previously shown to block the LPS-induced neutrophilic inflammation, the persistent increase in (f) seen at 24 h was attenuated, but neither compound was able to attenuate either the increase in (f) or the spontaneous bronchoconstriction seen at 2 h. In summary, the intra-tracheal LPS-challenge of rats results in pulmonary inflammation and dysfunction, which is similar to that seen in COPD patients. We conclude that the early increase in (f) and bronchoconstriction are not dependent upon airway inflammation, but airway inflammation most likely contributes to the persistent increase in (f) seen at 24 h.
Collapse
Affiliation(s)
- J Spond
- Schering Plough Research Institute, Allergy and Immunology, 2015 Galloping Hill Rd, K-15-1600, Kenilworth, NJ 07033, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Spond J, Case N, Chapman RW, Crawley Y, Egan RW, Fine J, Hey JA, Kreutner W, Kung T, Wang P, Minnicozzi M. Inhibition of experimental acute pulmonary inflammation by pirfenidone. Pulm Pharmacol Ther 2003; 16:207-14. [PMID: 12850123 DOI: 10.1016/s1094-5539(03)00026-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Pirfenidone, a putative tumor necrosis factor-alpha (TNF-alpha) inhibitor, has recently gained recognition for its therapeutic use in the treatment of idiopathic pulmonary fibrosis. As pulmonary fibrosis may be the result of lung inflammatory processes, we examined the anti-inflammatory potential of pirfenidone in several models of acute pulmonary inflammation. In antigen-induced allergic paradigms, 24 h after antigen challenge, sensitized mice or guinea pigs develop a prominent pulmonary inflammation, reflected by a significant increase in the number of recoverable bronchoalveolar lavage (BAL) total cells and eosinophils. In both species, the pretreatment of animals with pirfenidone (10 and 30 mg/kg) resulted in a dose-dependent inhibition of the antigen-induced pulmonary inflammation, which was reflected by a significant decrease in the BAL eosinophils and total cells by the 30 mg/kg dose. In a non-allergic model of pulmonary inflammation, rats challenged with intratracheal LPS develop a significant increase in BAL neutrophils and total cells, along with significant increases in TNF-alpha and IL-6. Pretreatment with pirfenidone (3 and 30 mg/kg) showed a dose-dependent inhibition of the LPS-induced pulmonary inflammation, reflected by a significant decrease in the number of BAL total and neutrophilic cells at both the 3 and 30 mg/kg dose. However, pirfenidone had no effect on the peak BAL levels of TNF-alpha. In contrast, pirfenidone significantly inhibited BAL levels of IL-6. In summary, we have shown that pirfenidone can inhibit allergic and non-allergic inflammatory cell recruitment and that its pulmonary anti-inflammatory activity is independent of TNF-alpha inhibition.
Collapse
Affiliation(s)
- J Spond
- Schering Plough Research Institute, Allergy and Immunology, 2015 Galloping Hill Road, K-15-1600, Kenilworth, NJ 07033, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kuss H, Hoefgen N, Johanssen S, Kronbach T, Rundfeldt C. In vivo efficacy in airway disease models of N-(3,5-dichloropyrid-4-yl)-[1-(4-fluorobenzyl)-5-hydroxy-indole-3-yl]-glyoxylic acid amide (AWD 12-281), a selective phosphodiesterase 4 inhibitor for inhaled administration. J Pharmacol Exp Ther 2003; 307:373-85. [PMID: 12944497 DOI: 10.1124/jpet.103.053942] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
N-(3,5-Dichloro-pyrid-4-yl)-[1-(4-fluorobenzyl)-5-hydroxy-indole-3-yl]-glyoxylic acid amide (AWD 12-281) is a highly potent and selective phosphodiesterase 4 (PDE4) inhibitor that was designed to have a metabolic profile that was optimized for topical administration. The aim of the current study was to explore the pharmacological profile of intratracheally administered AWD 12-281 in different models of asthma and chronic obstructive pulmonary disease (COPD) in comparison with steroids. To assess the anti-inflammatory potential of AWD 12-281, the antigen-induced cell infiltration in bronchoalveolar lavage fluid (BALF) of Brown Norway rats was determined. AWD 12-281 (ID50 of 7 microg/kg i.t.) as well as beclomethasone (0.1microg/kg i.t.) suppresses late-phase eosinophilia when administered intrapulmonary. Furthermore, AWD 12-281 has also strong anti-inflammatory properties when tested in lipopolysaccharide-induced acute lung neutrophilia in Lewis rats (ID50 of 0.02 microg/kg i.t.), ferrets (ID50 of 10 microg/kg i.t.), and domestic pigs (2-4 mg/pig i.t. or 1 mg/kg i.v.). In pigs, AWD 12-281 was as effective as beclomethasone (0.4 mg/pig i.t.) and dexamethasone (0.28 mg/kg i.v.), although at 3 to 10 times the dosage. The bronchodilatory activity of AWD 12-281 was assessed in sensitized guinea pigs. AWD 12-281 (1.5 mg/kg i.t., 1-h pretreatment) inhibited allergen-induced bronchoconstriction by 68% (parameter airway resistance). In sensitized BP-2 mice AWD 12-281 abolished the allergen-induced bronchial hyperresponsiveness and eosinophilia in BALF, showing dose dependence. When given orally, i.v. or i.t., AWD 12-281 has a considerably lower emetic potential than cilomilast in ferrets and roflumilast in pigs. When given topically by inhalation, no emesis could be induced in dogs up to the highest feasible dose (15 mg/kg in 50% lactose blend). These results indicate that AWD 12-281 is a unique potential new drug for the topical treatment of asthma and COPD.
Collapse
Affiliation(s)
- H Kuss
- Department of Pharmacology, elbion AG, Radebeul, Germany.
| | | | | | | | | |
Collapse
|
32
|
Affiliation(s)
- P J Barnes
- National Heart and Lung Institute, Imperial College School of Medicine, London SW3 6LY, UK.
| |
Collapse
|
33
|
Wang D, Deng C, Bugaj-Gaweda B, Kwan M, Gunwaldsen C, Leonard C, Xin X, Hu Y, Unterbeck A, De Vivo M. Cloning and characterization of novel PDE4D isoforms PDE4D6 and PDE4D7. Cell Signal 2003; 15:883-91. [PMID: 12834813 DOI: 10.1016/s0898-6568(03)00042-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We report here the cloning and characterization of two novel PDE4D isoforms, PDE4D6 and PDE4D7. PDE4D6 is a supershort form and PDE4D7 a long form of PDE4D. In addition, we have identified another novel long-form variant, PDE4D8, in silico. Like other isoforms, PDE4D6 and PDE4D7 are differentially expressed. Expression of PDE4D6 is restricted to brain whereas PDE4D7 is widely expressed in many tissues. Baculovirus-expressed recombinant PDE4D6 and PDE4D7 enzymes have high affinity for cyclic AMP (cAMP) and are inhibited by rolipram. The activity of PDE4D7, not PDE4D6, is elevated by a protein kinase A (PKA)-dependent mechanism, presumably through phosphorylation of the conserved PKA site in the upstream conserved region 1 (UCR1) domain. In agreement with early reports, human PDE4D6 and PDE4D7 are localized on genomic fragments of chromosome 5. Examination of the promoter regions reveals multiple CREB binding sites upstream of the starting methionine (Met) of each gene, suggesting that the cAMP/PKA signaling pathway may regulate transcriptional expression of PDE4D6 and PDE4D7.
Collapse
MESH Headings
- 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors
- 3',5'-Cyclic-AMP Phosphodiesterases/genetics
- 3',5'-Cyclic-AMP Phosphodiesterases/metabolism
- Alternative Splicing/genetics
- Amino Acid Sequence
- Animals
- Base Sequence
- Binding Sites/genetics
- Binding, Competitive/drug effects
- Blotting, Western
- Catalytic Domain/genetics
- Cell Line
- Cloning, Molecular
- Conserved Sequence/genetics
- Cyclic AMP/metabolism
- Cyclic AMP Response Element-Binding Protein/metabolism
- Cyclic AMP-Dependent Protein Kinases/pharmacology
- Cyclic Nucleotide Phosphodiesterases, Type 3
- Cyclic Nucleotide Phosphodiesterases, Type 4
- Dose-Response Relationship, Drug
- Gene Expression Regulation, Enzymologic
- Humans
- Isoenzymes/antagonists & inhibitors
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Kinetics
- Male
- Mice
- Molecular Sequence Data
- Phosphorylation
- Polymerase Chain Reaction
- Promoter Regions, Genetic/genetics
- Rats
- Rolipram/pharmacology
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Spodoptera
- Transfection
Collapse
Affiliation(s)
- Daguang Wang
- Discovery Research, Memory Pharmaceuticals Corp, 100 Philips Parkway, Montvale, NJ 07645, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Profita M, Chiappara G, Mirabella F, Di Giorgi R, Chimenti L, Costanzo G, Riccobono L, Bellia V, Bousquet J, Vignola AM. Effect of cilomilast (Ariflo) on TNF-alpha, IL-8, and GM-CSF release by airway cells of patients with COPD. Thorax 2003; 58:573-9. [PMID: 12832668 PMCID: PMC1746747 DOI: 10.1136/thorax.58.7.573] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Inflammation in chronic obstructive pulmonary disease (COPD) is characterised by increased neutrophilic infiltration of the airways. Cilomilast, a novel selective phosphodiesterase 4 inhibitor in clinical development for COPD treatment, exerts anti-inflammatory effects. The ability of cilomilast to inhibit the release of neutrophil chemoattractants such as tumour necrosis factor (TNF)-alpha, interleukin (IL)-8, and granulocyte-macrophage colony stimulating factor (GM-CSF) by bronchial epithelial cells and sputum cells isolated from 10 patients with COPD, 14 normal controls, and 10 smokers was investigated. METHODS Bronchial epithelial cells obtained by bronchial brushing and sputum cells isolated from induced sputum samples were cultured for 24 hours in the presence or absence of cilomilast (1 micro M). After incubation the supernatants were harvested and the levels of mediators measured by ELISA. Chemotactic activity in supernatants was also measured using a Boyden chamber. RESULTS TNF-alpha and IL-8 release by bronchial epithelial cells and sputum cells was higher in patients with COPD than in controls (p<0.0001) and smokers (p<0.0001). GM-CSF was only detectable in sputum cell supernatants and its level was higher in patients with COPD than in controls and smokers (p<0.0001, respectively). Cilomilast significantly reduced TNF-alpha release by bronchial epithelial cells and sputum cells (p=0.005) and GM-CSF release by sputum cells (p=0.003), whereas IL-8 release was not statistically inhibited. Supernatants of sputum cells and bronchial epithelial cells treated with cilomilast significantly decreased neutrophil chemotaxis (p<0.006 and p<0.008, respectively). CONCLUSIONS Cilomilast inhibits the production of some neutrophil chemoattractants by airway cells. This drug may play a role in the resolution of neutrophilic inflammation associated with COPD and cigarette smoke.
Collapse
Affiliation(s)
- M Profita
- Istituto di Medicina Generale e Pneumologia, Università di Palermo, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
COPD is one of the most common diseases in the world, and there is a global increase in prevalence, but there are no drugs available at present that halt the relentless progression of this disease. However, a better understanding of the cellular and molecular mechanisms that are involved in the underlying inflammatory and destructive processes has revealed several new targets for which drugs are now in development, and the prospects for finding new treatments are good.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College School of Medicine, Dovehouse Street, London SW3 6LY, UK.
| |
Collapse
|
36
|
Trifilieff A, Wyss D, Walker C, Mazzoni L, Hersperger R. Pharmacological profile of a novel phosphodiesterase 4 inhibitor, 4-(8-benzo[1,2,5]oxadiazol-5-yl-[1,7]naphthyridin-6-yl)-benzoic acid (NVP-ABE171), a 1,7-naphthyridine derivative, with anti-inflammatory activities. J Pharmacol Exp Ther 2002; 301:241-8. [PMID: 11907180 DOI: 10.1124/jpet.301.1.241] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We investigated the pharmacology of a new class of phosphodiesterase 4 (PDE4) inhibitor, 6,8-disubstituted 1,7-naphthyridines, by using 4-(8-benzo[1,2,5]oxadiazol-5-yl-[1,7]naphthyridin-6-yl)-benzoic acid (NVP-ABE171) as a representative compound and compared its potency with the most advanced PDE4 inhibitor, undergoing clinical trials, Ariflo [cis-4-cyano-4-(3-cyclopentyloxy-4-methoxyphenyl-r-1-cyclohexanecarboxylic acid)]. NVP-ABE171 inhibited the activity of phosphodiesterase 4A, 4B, 4C, and 4D with respective IC(50) values of 602, 34, 1230, and 1.5 nM. Ariflo was about 40 times less potent. In human cells, NVP-ABE171 inhibited the eosinophil and neutrophil oxidative burst, the release of cytokines by T cells, and the tumor necrosis factor-alpha release from monocytes, in the nanomolar range. Ariflo presented a similar inhibition profile but was 7 to 50 times less potent. In BALB/c mice challenged with lipopolysaccharide, NVP-ABE171 inhibited the airway neutrophil influx and activation with an ED(50) in the range of 3 mg/kg. Ariflo was inactive up to a dose of 10 mg/kg. In ovalbumin sensitized Brown Norway rats, NVP-ABE171 inhibited the lipopolysaccharide-induced airway neutrophil influx and activation (ED(50) of 0.2 mg/kg) and the ovalbumin-induced airway eosinophil influx and activation (ED(50) of 0.1 mg/kg). Ariflo was about 100 times less potent in both models. In the ovalbumin model, NVP-ABE171 had a duration of action of more than 24 h. NVP-ABE171 is a novel PDE4 inhibitor that shows activity both in vitro on human inflammatory cells and in vivo in animal models of lung inflammation. This compound class may have potential for the treatment of airway inflammatory conditions such as asthma and chronic obstructive pulmonary diseases.
Collapse
|