1
|
Cazzola M, Page C, Rogliani P, Calzetta L, Matera MG. PI3K Inhibitors as Potential Therapeutic Agents for the Treatment of COPD with Associated Atherosclerosis. Drugs 2025; 85:741-753. [PMID: 40214902 PMCID: PMC12098434 DOI: 10.1007/s40265-025-02179-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2025] [Indexed: 05/24/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) and cardiovascular disease (CVD) share a complex and multifactorial relationship characterized by overlapping risk factors, systemic inflammation, and intertwined pathophysiological mechanisms, with atherosclerosis emerging as a central inflammatory process connecting COPD and CVD, driven by systemic inflammation, oxidative stress, and endothelial dysfunction. While systemic inflammation is recognized as a critical link between these conditions, the precise pathways through which inflammation arises remain under investigation. There is therefore a need for therapeutic strategies to mitigate cardiovascular risks in patients with COPD. Among the pathways contributing to this interplay, the phosphoinositide 3-kinase (PI3K) signaling pathway has gained significant attention. Dysregulated PI3K signaling contributes to inflammation, oxidative stress, and endothelial dysfunction, which are key drivers of both COPD and CVD. Consequently, PI3K inhibitors have emerged as a promising therapeutic approach to mitigate inflammation and oxidative damage, offering a targeted strategy to address the shared pathological mechanisms underlying these diseases. A comprehensive understanding of the role of PI3K signaling and its inhibitors could facilitate the development of novel interventions to reduce cardiovascular risk in patients with COPD.
Collapse
Affiliation(s)
- Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Via Montpellier 1, 00131, Rome, Italy.
| | - Clive Page
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome 'Tor Vergata', Via Montpellier 1, 00131, Rome, Italy
| | - Luigino Calzetta
- Unit of Respiratory Diseases and Lung Function, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| |
Collapse
|
2
|
Yang Q, Tang, P, Zhang X. Effects of additional oral theophylline with inhaled therapy in patients with stable chronic obstructive pulmonary disease: A systematic review and meta-analysis. PLoS One 2025; 20:e0321984. [PMID: 40327637 PMCID: PMC12054895 DOI: 10.1371/journal.pone.0321984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 03/13/2025] [Indexed: 05/08/2025] Open
Abstract
The rationale for additional treatment of oral theophylline with inhaled therapy in patients with stable chronic obstructive pulmonary disease (COPD) is unclear. The databases including The Cochrane Library、PubMed、Embase and Web of Science were searched to collect randomized controlled trials (RCTs) involving the inhaled therapy plus additional theophylline therapy for the treatment of patients with stable COPD up to December 31, 2023. The forced expiratory volume in 1 second (FEV1)、forced expiratory volume in 1s% predicted (FEV1% pred)、forced vital capacity (FVC)、FEV1/FVC%、peak expiratory flow rate(PEFR)、exacerbation rate of COPD、COPD related hospital admissions、total symptom score and drug-related adverse reactions were extracted from literatures and the meta-analysis was conducted using the RevMan 5.4 software. 10 RCTs involving 2771 patients were included. The meta-analysis results showed that additional theophylline improved FEV1 with MD 0.08 (95% CI: 0.06 to 0.09, p<0.00001)、FVC with MD 0.13 (95% CI: 0.10 to 0.15, p<0.00001), reduce the risk of exacerbation rate with OR 0.75 (95% CI: 0.60 to 0.94, p=0.01) and COPD related hospital admissions with MD -0.07 (95% CI: -0.13 to -0.01, p=0.01). However, there was no significant difference in FEV1% pred with MD 0.45 (95% CI: -1.41 to 2.30, p=0.64)、FEV1/FVC% with MD -0.24 (95% CI: -3.26 to 2.79, p=0.88) and total symptom score with MD -0.03 (95% CI: -0.14 to 0.09, p=0.65). Furthermore, additional theophylline therapy induced a high incidence of drug-related adverse reactions with OR 1.33 (95% CI: 1.12 to 1.58, p=0.001), especially in gastrointestinal adverse reactions. Oral theophylline could be a supplementary therapeutic option when inhaled therapy is insufficient regarding of improvement in pulmonary function and reducing in exacerbation risk. However, additional theophylline therapy could increase the risk of drug-related adverse reactions and should be concerned.
Collapse
Affiliation(s)
- Qiang Yang
- Department of Pharmacy, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Pingxiu Tang,
- Department of Pharmacy, Suining Central Hospital, Suining City, Sichuan Province, China
| | - Xunyan Zhang
- Department of Pharmacy, Suining Central Hospital, Suining City, Sichuan Province, China
| |
Collapse
|
3
|
Baker JR, Daly L, Hassibi S, Kimura G, Nishimoto Y, Kizawa Y, Ito K. Senolytic therapy reduces inflammation in epithelial cells from COPD patients and in smoke-exposure mice. Front Med (Lausanne) 2025; 12:1451056. [PMID: 40357269 PMCID: PMC12066254 DOI: 10.3389/fmed.2025.1451056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Chronic obstructive pulmonary disease (COPD) is a disease of accelerated lung aging, with increased numbers of senescent cells found within the COPD Lung. Senescent cells may drive pathology by causing defective tissue repair and driving chronic inflammation via the release of inflammatory mediators known as the senescence-associated secretory phenotype (SASP). Senolytics are a new class of drugs that selectively remove senescent cells but have not previously been studied in COPD. We examined whether senescent cells are maintained during differentiation of COPD airway epithelial cells at the air-liquid interface and examined the role of the senolytic combination of dasatinib and quercetin on these cells and in a smoke-exposure mouse model. Methods Non-smoker and COPD bronchial epithelial cells were differentiated at air-liquid interface (ALI). Senescence markers (p16INKA and p21WAF1) were determined using Western blotting and SASP factors via Olink proteomics and Meso Scale Diagnostics (MSD). Cells and 11 days cigarette smoke (CS)-exposed mice were treated with the senolytic cocktail of dasatinib and quercetin (D + Q). Results Increased senescence markers were maintained in COPD ALI epithelium when differentiated at air-liquid interface, and treatment with D + Q reduced senescence markers, proteases, and Th2 cytokines. Therapeutic oral treatment of D + Q to CS-exposed mice reduced senescence burden while reducing inflammatory cell infiltrates and mouse CXCL1. Conclusion COPD subjects show increased airway epithelial senescence, and these cells can be cleared therapeutically using the senolytic cocktail of D + Q, reducing broad-spectrum pulmonary inflammation in vitro and in vivo.
Collapse
Affiliation(s)
- Jonathan R. Baker
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Leah Daly
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Shyreen Hassibi
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Genki Kimura
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Funabashi, Japan
| | - Yuki Nishimoto
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Funabashi, Japan
| | - Yasuo Kizawa
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Funabashi, Japan
| | - Kazuhiro Ito
- National Heart and Lung Institute, Imperial College, London, United Kingdom
| |
Collapse
|
4
|
Goyal A, Chopra V, Garg K, Sharma S. Mechanisms coupling the mTOR pathway to chronic obstructive pulmonary disease (COPD) pathogenesis. Cytokine Growth Factor Rev 2025; 82:55-69. [PMID: 39799015 DOI: 10.1016/j.cytogfr.2024.12.005] [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: 10/18/2024] [Revised: 12/10/2024] [Accepted: 12/26/2024] [Indexed: 01/15/2025]
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a poorly reversible respiratory disorder distinguished by dyspnea, cough, expectoration and exacerbations due to abnormality of airways or emphysema. In this review, we consider the therapeutic potential of targeting Mammalian target of Rapamycin (mTOR) for treating COPD. The mTOR is a highly conserved serine-threonine protein kinase that integrates signals from growth factors and nutrients to control protein synthesis, lipid biogenesis and metabolism. Dysregulated mTOR pathway signaling due to genetic factors or cigarette smoking impairs autophagy, driving the buildup of abnormal cells and damaged proteins, resulting in inflammation and oxidative stress. Persistent mTOR activation also contributes to pulmonary vascular cell proliferation, facilitating the development of pulmonary resistance in COPD. Rapamycin, an inhibitor of mTOR, prevents the buildup of senescent cells in the lungs of COPD patients and inhibits the release of lung tissue-damaging proteases. mTOR also impacts the corticosteroid sensitivity in COPD patients by regulating the levels of histone deacetylases. The emerging role of gut-lung axis dysbiosis in the progression of COPD and its influence on mTOR further highlights the relevance of the mTOR pathway in COPD pathophysiology.
Collapse
Affiliation(s)
- Ankita Goyal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Vishal Chopra
- Department of Pulmonary Medicine, Government Medical College, Patiala, India
| | - Kranti Garg
- Department of Pulmonary Medicine, Government Medical College, Patiala, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India.
| |
Collapse
|
5
|
To M, Arimoto Y, Honda N, Furusho N, Kinouchi T, Takeshita Y, Haruki K, To Y. Elevated oxidative stress and steroid insensitivity in patients with asthma and high body fat percentage. Ann Allergy Asthma Immunol 2025:S1081-1206(25)00126-7. [PMID: 40097096 DOI: 10.1016/j.anai.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Obesity is a risk factor for poor asthma control. Previous research suggests that patients with asthma and obesity have reduced responsiveness to corticosteroids. Recent studies indicate that body fat percentage may be more strongly associated with obesity-related diseases compared with body mass index. However, the relationship between body fat percentage and asthma, particularly regarding steroid sensitivity, remains unclear. OBJECTIVE To investigate the association between body fat percentage and steroid sensitivity in patients with asthma and elucidate the potential mechanisms underlying this association. METHODS Adult patients with asthma were enrolled and categorized into patients with high body fat percentage (HBF) and control groups. Peripheral blood mononuclear cells were isolated from the blood samples. These cells were cultured with dexamethasone followed by stimulation with tumor necrosis factor-α to assess the half-maximal inhibitory concentration of dexamethasone (IC50-Dex). Serum adipocytokines and oxidative stress markers were also measured. The effects of metformin on steroid sensitivity and oxidative stress in peripheral blood mononuclear cells were evaluated ex vivo. RESULTS The HBF group exhibited significantly higher IC50-Dex values than the control group. In the HBF group, IC50-Dex correlated with the number of acute exacerbations per year and serum oxidative stress marker levels. Treatment with metformin significantly reduced both IC50-Dex and oxidative stress marker levels in the HBF group. CONCLUSION Oxidative stress associated with increased body fat may contribute to impaired steroid sensitivity in patients with asthma. Metformin may improve steroid sensitivity by reducing oxidative stress, suggesting a potential therapeutic approach in this patient population.
Collapse
Affiliation(s)
- Masako To
- Department of Laboratory Medicine, Dokkyo Medical University, Saitama Medical Center, Saitama, Japan; Department of Respiratory Medicine, The Fraternity Memorial Hospital, Tokyo, Japan.
| | - Yoshihito Arimoto
- Department of Laboratory Medicine, Dokkyo Medical University, Saitama Medical Center, Saitama, Japan
| | - Natsue Honda
- Department of Laboratory Medicine, Dokkyo Medical University, Saitama Medical Center, Saitama, Japan
| | - Naho Furusho
- Department of Pulmonary Medicine, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Toru Kinouchi
- Department of Pulmonary Medicine, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Yuichiro Takeshita
- Department of Pulmonary Medicine, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Kosuke Haruki
- Department of Laboratory Medicine, Dokkyo Medical University, Saitama Medical Center, Saitama, Japan
| | - Yasuo To
- Department of Pulmonary Medicine, International University of Health and Welfare Narita Hospital, Chiba, Japan
| |
Collapse
|
6
|
Tahri S, Fakhfakh R, Bahloul E, Charfi S, Sellami K, Hachicha H, Boudawara T, Turki H, Masmoudi H, Abida O. Exploring the role of glucocorticoid receptors and co-chaperones in Pemphigus foliaceus stratification. Arch Dermatol Res 2025; 317:479. [PMID: 39988616 DOI: 10.1007/s00403-025-03864-1] [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: 10/19/2024] [Revised: 01/03/2025] [Accepted: 01/18/2025] [Indexed: 02/25/2025]
Abstract
Glucocorticoids (GC) are the main treatment for pemphigus foliaceus (PF). The effects of GC are mediated through glucocorticoid receptors (GR), with GRα and GRβ being the most significant isoforms. Several molecules are involved in mediating the cellular response to GC and can affect the response to treatment. However, the relationship between sensitivity to GC and the expression of GRα, GRβ, FKBP5, FKBP4, HAT1 and HDAC2 in PF disease has not yet been studied. The purpose of this study was to determine the expression of these molecules in patients with different types of response to treatment. Quantitative real-time PCR was used for gene expression profiling in systemic and cutaneous levels. The protein expression levels of GRα and GRβ and FKBP5 was accomplished through immunohistochemical staining. We studied the association of rs1360780 > FKBP5 SNP with PF disease using TaqMan SNP genotyping. Our findings showed downregulation in the gene expression levels of GRα (p = 0.016), HDAC2 (p = 0.004) and FKBP5 (p = 0.032) genes in PF patients compared to healthy controls in PBMC and an up-regulation in GRα (p = 0.041) and HAT1 (p = 0.008) in remittent patients compared to newly diagnosed patients in skin biopsies. GRα and GRβ proteins were less abundant at the cytoplasmic level in patients (p = 0.048 and 0.005, respectively). The nuclear score of FKBP5 was downregulated in patients (p = 0.028). Moreover, our results revealed that the rs1360780 > T allele is a risk factor to the endemic PF form. This study reported for the first-time the involvement of GRα, GRβ, FKBP5, FKBP4, HAT1 and HDAC2 in PF disease in Tunisian population.
Collapse
Affiliation(s)
- Safa Tahri
- Research Laboratory LR18/SP12 Auto-Immunity, Cancer and Immunogenetics, Immunology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, Tunisia.
| | - Raouia Fakhfakh
- Research Laboratory LR18/SP12 Auto-Immunity, Cancer and Immunogenetics, Immunology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, Tunisia
| | - Emna Bahloul
- Dermatology Department, Hedi Chaker Hospital, University of Sfax, Sfax, Tunisia
| | - Slim Charfi
- Pathology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, Tunisia
| | - Khadija Sellami
- Dermatology Department, Hedi Chaker Hospital, University of Sfax, Sfax, Tunisia
| | - Hend Hachicha
- Immunology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, Tunisia
| | - Tahya Boudawara
- Pathology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, Tunisia
| | - Hamida Turki
- Dermatology Department, Hedi Chaker Hospital, University of Sfax, Sfax, Tunisia
| | - Hatem Masmoudi
- Research Laboratory LR18/SP12 Auto-Immunity, Cancer and Immunogenetics, Immunology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, Tunisia
| | - Olfa Abida
- Research Laboratory LR18/SP12 Auto-Immunity, Cancer and Immunogenetics, Immunology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, Tunisia.
| |
Collapse
|
7
|
Manrui L, Xu Y, Liu J, Zhang X, Yuan R, Sun Y, Sun Y, Yang Q, Liao M, Lv M, Hu X, Chen X, Liang W. Aminophylline targets miR-128-3p/Slc7a11 axis to attenuate neuronal ferroptosis after traumatic brain injury. Cell Mol Life Sci 2025; 82:87. [PMID: 39985589 PMCID: PMC11846823 DOI: 10.1007/s00018-025-05601-3] [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: 09/02/2024] [Revised: 01/06/2025] [Accepted: 01/22/2025] [Indexed: 02/24/2025]
Abstract
Traumatic brain injury (TBI) is a significant global health issue, characterized by high rates of morbidity and mortality, along with substantial economic strains on healthcare systems. This study explores the potential of Aminophylline (AMP), a medication traditionally used for cardiovascular conditions and bronchiectasis, to enhance TBI outcomes by protecting against neuronal damage. Our findings indicate that AMP treatment significantly reduces neuronal ferroptosis in the cortex, leading to less tissue damage and notable improvements in cognitive and motor functions in mice subjected to controlled cortical impact (CCI). Additionally, we found that TBI resulted in decreased expression of miR-128-3p, a reduction that was further strengthened by AMP treatment. Gain-of-function experiments showed that overexpressing miR-128-3p increases neuronal ferroptosis by targeting Slc7a11, indicating how AMP mitigates cognitive and motor impairments in CCI mice. This study highlights the potential of AMP in treating TBI through the miR-128-3p/Slc7a11 pathway, marking the first report of its protective effects against ferroptosis in TBI.
Collapse
Affiliation(s)
- Li Manrui
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China
| | - Yang Xu
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China
| | - Jinyuan Liu
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China
| | - Xiao Zhang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, Sichuan, 637000, China
| | - Ruixuan Yuan
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China
| | - Yuwen Sun
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China
| | - Yihan Sun
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China
| | - Qiuyun Yang
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China
| | - Miao Liao
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China
| | - Meili Lv
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China
| | - Xin Hu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xiameng Chen
- Department of Forensic Pathology and Forensic Clinical Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China.
| | - Weibo Liang
- Department of Forensic Genetics, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Sichuan, 610000, Chengdu, China.
| |
Collapse
|
8
|
Bruno P, Pala D, Micoli A, Corsi M, Accetta A, Carzaniga L, Ronchi P, Fiorelli C, Formica M, Pizzirani D, Mazzucato R, Guariento S, Bertolini S, Martucci C, Allen AD, Mileo V, Capacchi S, Gallo PM, Fioni A, Xanxo Fernandez S, Villetti G, Puccini P, Civelli M, Guala M, Retini M, Martinelli P, Visentini F, Pavoni V, Daldosso M, Fontana S, Biagetti M, Capelli AM. Discovery of CHF-6523, an Inhaled Selective PI3Kδ Inhibitor for the Treatment of Chronic Obstructive Pulmonary Disease. J Med Chem 2025; 68:2444-2465. [PMID: 39635891 DOI: 10.1021/acs.jmedchem.4c02062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
The design of inhaled selective phosphatidylinositol 3-kinase delta (PI3Kδ) inhibitors for the treatment of inflammatory lung diseases was pursued. Knowledge-based design of a novel isocoumarin scaffold that was able to adopt a propeller-shape topology ensured the desired PI3Kδ selectivity. Achievement of low nanomolar cellular potencies through hinge binder group optimization, reduction of intrinsic permeability through head group optimization to extend lung retention, and screening of crystalline forms suitable for administration as dry powders culminated in the identification of compound 18. This novel inhaled selective PI3Kδ inhibitor displayed durable anti-inflammatory activity in a disease-relevant rat model of Th-2-driven acute lung inflammation and safe in vitro and in vivo preclinical profiles. Therefore, compound 18 showed the appropriate discovery profile and was progressed to clinical trials in healthy volunteers and chronic obstructive pulmonary disease (COPD) patients as CHF-6523.
Collapse
Affiliation(s)
- Paolo Bruno
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Daniele Pala
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Alessandra Micoli
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Mauro Corsi
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Alessandro Accetta
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Laura Carzaniga
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Paolo Ronchi
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Claudio Fiorelli
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Michele Formica
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Daniela Pizzirani
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Roberta Mazzucato
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Sara Guariento
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Serena Bertolini
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Cataldo Martucci
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Andrew Dennis Allen
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Valentina Mileo
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Silvia Capacchi
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Paola Maria Gallo
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Alessandro Fioni
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | | | - Gino Villetti
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Paola Puccini
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Maurizio Civelli
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Matilde Guala
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Michele Retini
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Prisca Martinelli
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Filippo Visentini
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Valentina Pavoni
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Matteo Daldosso
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Stefano Fontana
- Aptuit, an Evotec Company, Via Alessandro Fleming, 4, 37135 Verona, Italy
| | - Matteo Biagetti
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Anna Maria Capelli
- Chiesi Farmaceutici S.p.A, Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| |
Collapse
|
9
|
Ashique S, Mishra N, Mantry S, Garg A, Kumar N, Gupta M, Kar SK, Islam A, Mohanto S, Subramaniyan V. Crosstalk between ROS-inflammatory gene expression axis in the progression of lung disorders. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:417-448. [PMID: 39196392 DOI: 10.1007/s00210-024-03392-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/16/2024] [Indexed: 08/29/2024]
Abstract
A significant number of deaths and disabilities worldwide are brought on by inflammatory lung diseases. Many inflammatory lung disorders, including chronic respiratory emphysema, resistant asthma, resistance to steroids, and coronavirus-infected lung infections, have severe variants for which there are no viable treatments; as a result, new treatment alternatives are needed. Here, we emphasize how oxidative imbalance contributes to the emergence of provocative lung problems that are challenging to treat. Endogenic antioxidant systems are not enough to avert free radical-mediated damage due to the induced overproduction of ROS. Pro-inflammatory mediators are then produced due to intracellular signaling events, which can harm the tissue and worsen the inflammatory response. Overproduction of ROS causes oxidative stress, which causes lung damage and various disease conditions. Invasive microorganisms or hazardous substances that are inhaled repeatedly can cause an excessive amount of ROS to be produced. By starting signal transduction pathways, increased ROS generation during inflammation may cause recurrent DNA damage and apoptosis and activate proto-oncogenes. This review provides information about new targets for conducting research in related domains or target factors to prevent, control, or treat such inflammatory oxidative stress-induced inflammatory lung disorders.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, 713212, India.
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| | - Neeraj Mishra
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University Madhya Pradesh (AUMP), Gwalior, MP, 474005, India
| | - Shubhrajit Mantry
- Department of Pharmaceutics, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Ashish Garg
- Department of Pharmaceutics, Guru Ramdas Khalsa Institute of Science and Technology (Pharmacy), Jabalpur, Madhya Pradesh, 483001, India
| | - Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to Be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh, 201204, India
| | - Madhu Gupta
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Delhi, 110017, India
| | - Sanjeeb Kumar Kar
- Department of Pharmaceutical Chemistry, Department of Pharmacy, Sarala Birla University, Ranchi, Jharkhand, 835103, India
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, Uttar Pradesh, 226026, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, Karnataka, 575018, India.
| | - Vetriselvan Subramaniyan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
10
|
Zeng XX, Gao DD, Zhang F. Cortex Mori Radicis[Morus Alba L. (Moraceae)] extract alleviates senescence via PI3K/Akt signaling in COPD fibroblasts. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156004. [PMID: 39326135 DOI: 10.1016/j.phymed.2024.156004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is marked by prolonged exposure to cigarette smoke, which accelerates senescence in lung fibroblasts and contributes to lung fibrosis. Cortex Mori Radicis [Morus albal. (Moraceae)], a traditional Chinese medicinal herb known for its antitussive properties, has emerged as a potential therapeutic agent for COPD. This study aims to elucidate the immunological mechanisms by which Cortex Mori Radicis mitigates COPD progression, utilizing a mouse model and the MRC-5 cell line. METHODS AND RESULTS COPD mouse models were established through chronic cigarette smoke (CS) exposure, followed by isolation of lung fibroblasts. Senescence markers and inflammatory mediators were assessed in both the isolated cells and the mice. Lung fibroblasts and bleomycin (Bleomycin)-treated MRC-5 cells exhibited elevated expression of senescence markers, including senescence-associated beta-galactosidase activity, p16INK4A, p21, p38 MAPK, and p53, along with increased levels of senescence-associated secretory phenotype (SASP) mRNA, such as IL-6 and IL-8. Treatment with Cortex Mori Radicis significantly attenuated the protein levels of these senescence markers and reduced SASP mRNA expression. Furthermore, integration of transcriptomic data from lung tissues and primary fibroblasts, combined with network pharmacology analysis, indicated that Cortex Mori Radicis inhibits fibroblast senescence via the PI3K/Akt pathway, thereby ameliorating lung pathology in COPD mice. CONCLUSION Through the application of transcriptomics and network analysis, this study identifies that Cortex Mori Radicis suppresses cigarette smoke-induced senescence in pulmonary tissues and bleomycin (Bleomycin)-exposed MRC-5 cells by targeting the PI3K/Akt signaling pathway. These findings underscore the therapeutic potential of Cortex Mori Radicis as a novel intervention for COPD.
Collapse
Affiliation(s)
- Xi-Xi Zeng
- The Joint Innovation Center for Health & Medicine, the Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China.
| | - Dan-Dan Gao
- Wenzhou Medical University, Wenzhou 325000, China
| | - Feng Zhang
- Department of Respiratory and Critical Care, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China; Joint Innovation Center for Engineering in Medicine, Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou 324000, China.
| |
Collapse
|
11
|
Bruno P, Micoli A, Corsi M, Pala D, Guariento S, Fiorelli C, Ronchi P, Fioni A, Gallo PM, Marenghi G, Bertolini S, Capacchi S, Mileo V, Biagetti M, Capelli AM. Discovery and Optimization of Pyridazinones as PI3Kδ Selective Inhibitors for Administration by Inhalation. J Med Chem 2024; 67:11103-11124. [PMID: 38907711 DOI: 10.1021/acs.jmedchem.4c00610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
A hit-to-lead campaign pursuing the identification of novel inhalant small-molecule phosphatidylinositol 3-kinase (PI3K) inhibitors for the treatment of inflammatory respiratory diseases is disclosed. A synthetically versatile pyridazin-3(2H)-one scaffold was designed, and three exit vectors on the core moiety were used to explore chemical diversity and optimize pharmacological and absorption, distribution, metabolism, and excretion (ADME) properties. Desired modulation of PI3Kδ selectivity and cellular potency as well as ADME properties in view of administration by inhalation was achieved. Intratracheal administration of lead compound 26 resulted in a promising pharmacokinetic profile, thus demonstrating that the optimization strategy of in vitro profiles successfully translated to an in vivo setting.
Collapse
Affiliation(s)
- Paolo Bruno
- Medicinal Chemistry and Drug Design Technologies Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Alessandra Micoli
- Drug Discovery Chemistry Department, Aptuit, an Evotec Company, Via A. Fleming 4, 37135 Verona, Italy
| | - Mauro Corsi
- In Vitro Biology Department, Aptuit, an Evotec Company, Via A. Fleming 4, 37135 Verona, Italy
| | - Daniele Pala
- Medicinal Chemistry and Drug Design Technologies Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Sara Guariento
- Medicinal Chemistry and Drug Design Technologies Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Claudio Fiorelli
- Medicinal Chemistry and Drug Design Technologies Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Paolo Ronchi
- Medicinal Chemistry and Drug Design Technologies Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Alessandro Fioni
- Pharmacokinetics Biochemistry Metabolism Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Paola Maria Gallo
- Pharmacokinetics Biochemistry Metabolism Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Giulia Marenghi
- Pharmacokinetics Biochemistry Metabolism Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Serena Bertolini
- Pharmacology Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Silvia Capacchi
- Medicinal Chemistry and Drug Design Technologies Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Valentina Mileo
- Analytics & Early Formulations Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Matteo Biagetti
- Pipeline Innovation Department, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| | - Anna Maria Capelli
- AIR Franchise, Chiesi Farmaceutici S.p.A, Nuovo Centro Ricerche, Largo Belloli 11/a, 43122 Parma, Italy
| |
Collapse
|
12
|
Balde A, Ramya CS, Nazeer RA. A review on current advancement in zebrafish models to study chronic inflammatory diseases and their therapeutic targets. Heliyon 2024; 10:e31862. [PMID: 38867970 PMCID: PMC11167310 DOI: 10.1016/j.heliyon.2024.e31862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/02/2024] [Accepted: 05/22/2024] [Indexed: 06/14/2024] Open
Abstract
Chronic inflammatory diseases are caused due to prolonged inflammation at a specific site of the body. Among other inflammatory diseases, bacterial meningitis, chronic obstructive pulmonary disease (COPD), atherosclerosis and inflammatory bowel diseases (IBD) are primarily focused on because of their adverse effects and fatality rates around the globe in recent times. In order to come up with novel strategies to eradicate these diseases, a clear understanding of the mechanisms of the diseases is needed. Similarly, detailed insight into the mechanisms of commercially available drugs and potent lead compounds from natural sources are also important to establish efficient therapeutic effects. Zebrafish is widely accepted as a model to study drug toxicity and the pharmacokinetic effects of the drug. Moreover, researchers use various inducers to trigger inflammatory cascades and stimulate physiological changes in zebrafish. The effect of these inducers contrasts with the type of zebrafish used in the investigation. Hence, a thorough analysis is required to study the current advancements in the zebrafish model for chronic inflammatory disease suppression. This review presents the most common inflammatory diseases, commercially available drugs, novel therapeutics, and their mechanisms of action for disease suppression. The review also provides a detailed description of various zebrafish models for these diseases. Finally, the future prospects and challenges for the same are described, which can help the researchers understand the potency of the zebrafish model and its further exploration for disease attenuation.
Collapse
Affiliation(s)
- Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Cunnathur Saravanan Ramya
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| |
Collapse
|
13
|
Park HJ, Lee JU, Jeon S, Lee HS, Kim BY, Chae YJ, Kim GO, Park JW, Lee JH. Prescription patterns and effectiveness of medications for chronic obstructive pulmonary disease: A retrospective study of real-world settings. PLoS One 2024; 19:e0304362. [PMID: 38857214 PMCID: PMC11164367 DOI: 10.1371/journal.pone.0304362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 05/11/2024] [Indexed: 06/12/2024] Open
Abstract
This study aimed to define real-world prescription patterns in Korea and compare the effectiveness of chronic obstructive pulmonary disease (COPD) medications. We used national claims data provided by the Health Insurance Review and Assessment Service in Korea and examined patients who were first diagnosed with COPD and started treatment between May 1, 2017, and April 30, 2018, with no change in drug regimen. Among 30,784 patients with COPD, long-acting β2 agonist (LABA) combined with long-acting muscarinic antagonist (LAMA) (32.7%), inhaled corticosteroid-LABA (ICS-LABA) (25.6%), LAMA (18.3%), ICS (5.8%), or LABA (4.6%) were prescribed as the first-choice inhalers. The use of LABA-LAMA (hazard ratio [HR], 0.248-0.584), LAMA (HR, 0.320-0.641), ICS-LABA (HR, 0.325-0.643), and xanthine (HR, 0.563-0.828) significantly reduced the total and severe exacerbation rates compared with no use of each medication. However, the use of ICS or LABA individually did not yield such effects. The continued use of LABA-LAMA, LAMA, and ICS-LABA showed a significant effect on exacerbation rate, whereas the long-term use of ICS, LABA, and xanthine did not. Moreover, some high doses of ICS-LABA did not show significant effects. This real-world study revealed that LAMA and/or LABA could be the first choice of therapy, as recommended by recent guidelines. However, ICS, xanthine, and high-dose ICS-LABA are still being prescribed frequently as first-line drugs in Korea.
Collapse
Affiliation(s)
- Hye Jung Park
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Uk Lee
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soyoung Jeon
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Bo Yeon Kim
- Healthcare Insurance Review & Assessment Service, Wonju, Republic of Korea
| | - Yu Jin Chae
- Healthcare Insurance Review & Assessment Service, Wonju, Republic of Korea
| | - Gui Ok Kim
- Healthcare Insurance Review & Assessment Service, Wonju, Republic of Korea
| | - Jung-Won Park
- Division of Allergy and Immunology, Department of Internal Medicine, Republic of Korea
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Hyun Lee
- Division of Allergy and Immunology, Department of Internal Medicine, Republic of Korea
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
14
|
Darlami O, Pun R, Ahn SH, Kim SH, Shin D. Macrocyclization strategy for improving candidate profiles in medicinal chemistry. Eur J Med Chem 2024; 272:116501. [PMID: 38754142 DOI: 10.1016/j.ejmech.2024.116501] [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: 04/06/2024] [Revised: 05/12/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
Macrocycles are defined as cyclic compounds with 12 or more members. In medicinal chemistry, they are categorized based on their core chemistry into cyclic peptides and macrocycles. Macrocycles are advantageous because of their structural diversity and ability to achieve high affinity and selectivity towards challenging targets that are often not addressable by conventional small molecules. The potential of macrocyclization to optimize drug-like properties while maintaining adequate bioavailability and permeability has been emphasized as a key innovation in medicinal chemistry. This review provides a detailed case study of the application of macrocyclization over the past 5 years, starting from the initial analysis of acyclic active compounds to optimization of the resulting macrocycles for improved efficacy and drug-like properties. Additionally, it illustrates the strategic value of macrocyclization in contemporary drug discovery efforts.
Collapse
Affiliation(s)
- Om Darlami
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon, 21935, Republic of Korea
| | - Rabin Pun
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon, 21935, Republic of Korea
| | - Sung-Hoon Ahn
- College of Pharmacy, Kangwon National University, Gangwondaehak-gil 1, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Seok-Ho Kim
- College of Pharmacy, Kangwon National University, Gangwondaehak-gil 1, Chuncheon, Gangwon-do, 24341, Republic of Korea.
| | - Dongyun Shin
- College of Pharmacy, Gachon University, Hambakmoe-ro 191, Yeunsu-gu, Incheon, 21935, Republic of Korea.
| |
Collapse
|
15
|
Scurek M, Brat K. A narrative review of theophylline: is there still a place for an old friend? J Thorac Dis 2024; 16:3450-3460. [PMID: 38883616 PMCID: PMC11170423 DOI: 10.21037/jtd-23-1781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 05/17/2024] [Indexed: 06/18/2024]
Abstract
Background and Objective Theophylline has been used for decades in human medicine for its psychostimulant, anti-inflammatory, and bronchodilator effects. Historically, in pulmonary medicine, theophylline has been used in the treatment of obstructive pulmonary diseases such as bronchial asthma (BA) or chronic obstructive pulmonary disease (COPD). This review aims to determine whether theophylline still has its place in the therapy of obstructive pulmonary diseases or whether we can even extend its use to other diagnoses such as atropine-resistant cardiac arrests, apnea of prematurity, or others. Moreover, we also aim to determine if there is a rationale for using low-dose theophylline due to its immunomodulatory and anti-inflammatory effect, or if the future of methylxanthines lies in newly synthesized derivates of theophylline such as bamifylline, or doxofylline. Methods The narrative review is based on a literature search of the articles indexed in the PubMed database in 2023. We searched the database since the year 2009 using the MeSH terms "theophylline", "aminophylline", and "methylxanthines" and we included original articles in the English language. Key Content and Findings Theophylline has a number of adverse drug reactions (ADRs), the most serious of which is its effect on the cardiovascular system. It can cause severe arrhythmias or even cardiac arrest when overdosed. On the other hand, there is still a substantial amount of its applications in current clinical practice. Conclusions There is considerable controversy associated with its use in current medicine, which can be attributed both to its narrow therapeutic range and its mentioned cardiotoxic effect. Herein, we summarize the current state-of-art of theophylline and its use in human medicine.
Collapse
Affiliation(s)
- Martin Scurek
- Department of Respiratory Diseases, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Kristian Brat
- Department of Respiratory Diseases, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| |
Collapse
|
16
|
Li Q, Jiang G, Lv Y. Inhibition of phosphoinositide 3-kinase activity attenuates neutrophilic airway inflammation and inhibits pyrin domain-containing 3 inflammasome activation in an ovalbumin-lipopolysaccharide-induced asthma murine model. Mol Biol Rep 2024; 51:698. [PMID: 38811424 PMCID: PMC11136729 DOI: 10.1007/s11033-024-09360-5] [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: 01/24/2024] [Accepted: 02/16/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Existing investigations suggest that the blockade of phosphoinositide 3-kinase (PI3K) activity contributes to inflammatory solution in allergic asthma, but whether this inhibition directly attenuates neutrophilic airway inflammation in vivo is still unclear. We explored the pharmacological effects of LY294002, a specific inhibitor of PI3K on the progression of neutrophilic airway inflammation and investigated the underlying mechanism. METHODS AND RESULTS Female C57BL/6 mice were intranasally sensitized with ovalbumin (OVA) together with lipopolysaccharide (LPS) on days 0 and 6, and challenged with OVA on days 14-17 to establish a neutrophilic airway disease model. In the challenge phase, a subset of mice was treated intratracheally with LY294002. We found that treatment of LY294002 attenuates clinic symptoms of inflammatory mice. Histological studies showed that LY294002 significantly inhibited inflammatory cell infiltration and mucus production. The treatment also significantly inhibited OVA-LPS induced increases in inflammatory cell counts, especially neutrophil counts, and IL-17 levels in bronchoalveolar lavage fluid (BALF). LY294002 treated mice exhibited significantly increased IL-10 levels in BALF compared to the untreated mice. In addition, LY294002 reduced the plasma concentrations of IL-6 and IL-17. The anti-inflammatory effects of LY29402 were correlated with the downregulation of NLRP3 inflammasome. CONCLUSIONS Our findings suggested that LY294002 as a potential pharmacological target for neutrophilic airway inflammation.
Collapse
Affiliation(s)
- Qun Li
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Bengbu Medical University, Anhui, China
| | - Guiyun Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China
| | - Yunxiang Lv
- Department of Pulmonary and Critical Care MedicineAnhui Clinical and Preclinical Key Laboratory of Respiratory DiseaseMolecular Diagnosis Center, The First Affiliated Hospital of Bengbu Medical University, Bengbu, 233000, Anhui, China.
| |
Collapse
|
17
|
Kimura G, Tagami A, Fukui R, Yaita M, Miyasaka T. Airway inflammation in a novel mouse model of asthma-COPD overlap induced by co-exposure to papain and tobacco smoke. Biochem Biophys Res Commun 2024; 709:149831. [PMID: 38552552 DOI: 10.1016/j.bbrc.2024.149831] [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: 02/15/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) are respiratory diseases associated with airway inflammation, which is the main pathogenesis. Although their causes and characteristics differ, in some cases, asthma and COPD may coexist in the same patient in a condition called asthma-COPD overlap (ACO). The prognosis of ACO is more unfavourable than those of asthma or COPD alone, without any treatment strategies demonstrating efficacy. Owing to its intricate spectrum of features, the detailed pathogenesis of how ACO exacerbates respiratory features remains unclear. In this study, we exposed papain-induced asthma model mice to tobacco smoke to establish an ACO mouse model, in which features of airway inflammation observed in both asthma and COPD were incorporated. This model exhibited distinctive mixed and corticosteroid-resistant airway inflammation and emphysematous changes that are characteristic of ACO. The novel mouse model established here is expected to significantly contribute to elucidating the mechanisms of the broad pathologies of ACO and identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Genki Kimura
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Funabashi, Japan.
| | - Ai Tagami
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Funabashi, Japan
| | - Rina Fukui
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Funabashi, Japan
| | - Masaki Yaita
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Funabashi, Japan
| | - Tomohiro Miyasaka
- Department of Physiology and Anatomy, Nihon University School of Pharmacy, Funabashi, Japan.
| |
Collapse
|
18
|
Roth‐Walter F, Adcock IM, Benito‐Villalvilla C, Bianchini R, Bjermer L, Caramori G, Cari L, Chung KF, Diamant Z, Eguiluz‐Gracia I, Knol EF, Jesenak M, Levi‐Schaffer F, Nocentini G, O'Mahony L, Palomares O, Redegeld F, Sokolowska M, Van Esch BCAM, Stellato C. Metabolic pathways in immune senescence and inflammaging: Novel therapeutic strategy for chronic inflammatory lung diseases. An EAACI position paper from the Task Force for Immunopharmacology. Allergy 2024; 79:1089-1122. [PMID: 38108546 PMCID: PMC11497319 DOI: 10.1111/all.15977] [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: 09/13/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
The accumulation of senescent cells drives inflammaging and increases morbidity of chronic inflammatory lung diseases. Immune responses are built upon dynamic changes in cell metabolism that supply energy and substrates for cell proliferation, differentiation, and activation. Metabolic changes imposed by environmental stress and inflammation on immune cells and tissue microenvironment are thus chiefly involved in the pathophysiology of allergic and other immune-driven diseases. Altered cell metabolism is also a hallmark of cell senescence, a condition characterized by loss of proliferative activity in cells that remain metabolically active. Accelerated senescence can be triggered by acute or chronic stress and inflammatory responses. In contrast, replicative senescence occurs as part of the physiological aging process and has protective roles in cancer surveillance and wound healing. Importantly, cell senescence can also change or hamper response to diverse therapeutic treatments. Understanding the metabolic pathways of senescence in immune and structural cells is therefore critical to detect, prevent, or revert detrimental aspects of senescence-related immunopathology, by developing specific diagnostics and targeted therapies. In this paper, we review the main changes and metabolic alterations occurring in senescent immune cells (macrophages, B cells, T cells). Subsequently, we present the metabolic footprints described in translational studies in patients with chronic asthma and chronic obstructive pulmonary disease (COPD), and review the ongoing preclinical studies and clinical trials of therapeutic approaches aiming at targeting metabolic pathways to antagonize pathological senescence. Because this is a recently emerging field in allergy and clinical immunology, a better understanding of the metabolic profile of the complex landscape of cell senescence is needed. The progress achieved so far is already providing opportunities for new therapies, as well as for strategies aimed at disease prevention and supporting healthy aging.
Collapse
Affiliation(s)
- F. Roth‐Walter
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine ViennaMedical University Vienna and University ViennaViennaAustria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and ImmunologyMedical University of ViennaViennaAustria
| | - I. M. Adcock
- Molecular Cell Biology Group, National Heart & Lung InstituteImperial College LondonLondonUK
| | - C. Benito‐Villalvilla
- Department of Biochemistry and Molecular Biology, School of ChemistryComplutense University of MadridMadridSpain
| | - R. Bianchini
- Comparative Medicine, The Interuniversity Messerli Research Institute of the University of Veterinary Medicine ViennaMedical University Vienna and University ViennaViennaAustria
| | - L. Bjermer
- Department of Respiratory Medicine and Allergology, Lung and Allergy research, Allergy, Asthma and COPD Competence CenterLund UniversityLundSweden
| | - G. Caramori
- Department of Medicine and SurgeryUniversity of ParmaPneumologiaItaly
| | - L. Cari
- Department of Medicine, Section of PharmacologyUniversity of PerugiaPerugiaItaly
| | - K. F. Chung
- Experimental Studies Medicine at National Heart & Lung InstituteImperial College London & Royal Brompton & Harefield HospitalLondonUK
| | - Z. Diamant
- Department of Respiratory Medicine and Allergology, Institute for Clinical ScienceSkane University HospitalLundSweden
- Department of Respiratory Medicine, First Faculty of MedicineCharles University and Thomayer HospitalPragueCzech Republic
- Department of Clinical Pharmacy & PharmacologyUniversity Groningen, University Medical Center Groningen and QPS‐NLGroningenThe Netherlands
| | - I. Eguiluz‐Gracia
- Allergy UnitHospital Regional Universitario de Málaga‐Instituto de Investigación Biomédica de Málaga (IBIMA)‐ARADyALMálagaSpain
| | - E. F. Knol
- Departments of Center of Translational Immunology and Dermatology/AllergologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - M. Jesenak
- Department of Paediatrics, Department of Pulmonology and Phthisiology, Comenius University in Bratislava, Jessenius Faculty of Medicine in MartinUniversity Teaching HospitalMartinSlovakia
| | - F. Levi‐Schaffer
- Institute for Drug Research, Pharmacology Unit, Faculty of MedicineThe Hebrew University of JerusalemJerusalemIsrael
| | - G. Nocentini
- Department of Medicine, Section of PharmacologyUniversity of PerugiaPerugiaItaly
| | - L. O'Mahony
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of MedicineUniversity College CorkCorkIreland
- School of MicrobiologyUniversity College CorkCorkIreland
| | - O. Palomares
- Department of Biochemistry and Molecular Biology, School of ChemistryComplutense University of MadridMadridSpain
| | - F. Redegeld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of ScienceUtrecht UniversityUtrechtThe Netherlands
| | - M. Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZürichDavosSwitzerland
- Christine Kühne – Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - B. C. A. M. Van Esch
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of ScienceUtrecht UniversityUtrechtThe Netherlands
| | - C. Stellato
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”University of SalernoSalernoItaly
| |
Collapse
|
19
|
Yang H, He S, Liang L, Pan J. Efficacy of Nemiralisib in Chronic Obstructive Pulmonary Disease: A Systematic Review. Clin Ther 2024; 46:360-367. [PMID: 38503629 DOI: 10.1016/j.clinthera.2024.02.008] [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: 11/08/2023] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 03/21/2024]
Abstract
PURPOSE Chronic obstructive pulmonary disease (COPD) is a major public health concern. Exacerbation of COPD leads to poor health and frequent episodes of increased systemic and airway inflammation. Immunomodulatory drugs have garnered extensive attention because they may reduce the rate of COPD exacerbation. This review aimed to evaluate the efficacy and safety of nemiralisib in COPD patients. METHODS Medical databases, including the Cochrane Library, EMBASE, and PubMed, were queried from inception to June 2023 to identify randomized controlled trials (RCTs) on the efficacy of nemiralisib in COPD patients. This systematic review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. The Cochrane Collaboration tool was used to assess the risk of bias of the included RCTs. Two authors independently conducted literature screening and data extraction. Key information from the included studies was extracted, tabulated, and compared using a data extraction table. Moreover, the key characteristics, quality, potential bias, and endpoint outcomes of the included studies were summarized. A meta-analysis was conducted when the study outcomes were sufficiently comparable, and the required data were available for extraction. FINDINGS Initially, 48 references were identified, leading to the inclusion of four trials. No significant difference was found between the nemiralisib and placebo groups in St George's Respiratory Questionnaire score, modified Medical Research Council Dyspnea Scale score, COPD Assessment Test score, time to next on-treatment exacerbation, proportion of patients achieving exacerbation recovery, time to exacerbation recovery, and rescue medication use. Contrastingly, the results demonstrated that nemiralisib may lower oral corticosteroid use during acute exacerbation of COPD. Meanwhile, the efficacy of nemiralisib on the exacerbation rate, as well as several parameters associated with lung function, including forced expiratory volume in 1 second, specific airway conductance, specific imaging airway wall thickness, distal specific imaging airway volume measured at functional residual capacity, specific imaging airway resistance, low attenuation score, and internal airflow lobar distribution in the lower pulmonary region, were conflicting. Attributed to the limited number of included RCTs and insufficient extracted data, it was not feasible to conduct a comprehensive meta-analysis. IMPLICATIONS Because of insufficient data, this systematic review could not make any definitive statement regarding the efficacy of nemiralisib in COPD patients. In terms of safety, nemiralisib was generally well tolerated. Further trials are required to explore the efficacy of this drug.
Collapse
Affiliation(s)
- Hongkuan Yang
- Respiratory Intensive Care Unit, Gaozhou People's Hospital, Maoming, Guangdong Province, China
| | - Shuifeng He
- The first section of the Department of Pulmonary and Critical Care Medicine, Gaozhou People's Hospital, Maoming, Guangdong Province, China
| | - Linbao Liang
- Respiratory Intensive Care Unit, Gaozhou People's Hospital, Maoming, Guangdong Province, China
| | - Junjie Pan
- Department of Pulmonary and Critical Care Medicine, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
20
|
Liu Y, Kong H, Cai H, Chen G, Chen H, Ruan W. Progression of the PI3K/Akt signaling pathway in chronic obstructive pulmonary disease. Front Pharmacol 2023; 14:1238782. [PMID: 37799975 PMCID: PMC10548138 DOI: 10.3389/fphar.2023.1238782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/08/2023] [Indexed: 10/07/2023] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a chronic respiratory disease characterized by a slow progression and caused by the inhalation of harmful particulate matter. Cigarette smoke and air pollutants are the primary contributing factors. Currently, the pathogenesis of COPD remains incompletely understood. The PI3K/Akt signaling pathway has recently emerged as a critical regulator of inflammation and oxidative stress response in COPD, playing a pivotal role in the disease's progression and treatment. This paper reviews the association between the PI3K/Akt pathway and COPD, examines effective PI3K/Akt inhibitors and novel anti-COPD agents, aiming to identify new therapeutic targets for clinical intervention in this disease.
Collapse
Affiliation(s)
- Yanhui Liu
- Department of Clinical Pharmacy, Anhui Provincial Children’s Hospital, Hefei, Anhui, China
| | - Haobo Kong
- Department of Respiratory Intensive Care Unit, Anhui Chest Hospital, Hefei, Anhui, China
| | - Heping Cai
- Department of Clinical Pharmacy, Anhui Provincial Children’s Hospital, Hefei, Anhui, China
| | - Guanru Chen
- Department of Clinical Pharmacy, Anhui Provincial Children’s Hospital, Hefei, Anhui, China
| | - Huiying Chen
- Department of Clinical Pharmacy, Anhui Provincial Children’s Hospital, Hefei, Anhui, China
| | - Wenyi Ruan
- Department of Clinical Pharmacy, Anhui Provincial Children’s Hospital, Hefei, Anhui, China
| |
Collapse
|
21
|
Tang Y, Zheng F, Bao X, Zheng Y, Hu X, Lou S, Zhao H, Cui S. Discovery of Highly Selective and Orally Bioavailable PI3Kδ Inhibitors with Anti-Inflammatory Activity for Treatment of Acute Lung Injury. J Med Chem 2023; 66:11905-11926. [PMID: 37606563 DOI: 10.1021/acs.jmedchem.3c00508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
PI3Kδ is a promising target for the treatment of inflammatory disease; however, the application of PI3Kδ inhibitors in acute respiratory inflammatory diseases is rarely investigated. In this study, through scaffold hopping design, we report a new series of 1H-pyrazolo[3,4-d]pyrimidin-4-amine-tethered 3-methyl-1-aryl-1H-indazoles as highly selective and potent PI3Kδ inhibitors with significant anti-inflammatory activities for treatment of acute lung injury (ALI). There were 29 compounds designed, prepared, and subjected to PI3Kδ inhibitory activity evaluation and anti-inflammatory activity evaluation in macrophages. (S)-29 was identified as a candidate with high PI3Kδ inhibitory activity, isoform selectivity, and high oral bioavailability. The in vivo administration of (S)-29 at 10 mg/kg dosage could significantly ameliorate histopathological changes and attenuate lung inflammation in lung tissues of LPS-challenged mice. Molecular docking demonstrated the success of scaffold hopping design. Overall, (S)-29 is a potent PI3Kδ inhibitor which might be a promising candidate for the treatment of ALI.
Collapse
Affiliation(s)
- Yongmei Tang
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fanli Zheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Xiaodong Bao
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yanan Zheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Xueping Hu
- Institute of Molecular Sciences and Engineering, Institute of Frontier and Interdisciplinary Science, Shandong University, Qingdao 266237, China
| | - Siyue Lou
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Huajun Zhao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311402, China
| | - Sunliang Cui
- Institute of Drug Discovery and Design, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
22
|
Ouyang L, Su G, Quan J, Xiong Z, Lai T. Emerging roles and therapeutic implications of HDAC2 and IL-17A in steroid-resistant asthma. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:108-112. [PMID: 39170824 PMCID: PMC11332885 DOI: 10.1016/j.pccm.2023.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Indexed: 08/23/2024]
Abstract
Steroid resistance represents a major clinical problem in the treatment of severe asthma, and therefore a better understanding of its pathogenesis is warranted. Recent studies indicated that histone deacetylase 2 (HDAC2) and interleukin 17A (IL-17A) play important roles in severe asthma. HDAC2 activity is reduced in patients with severe asthma and smoking-induced asthma, perhaps accounting for the amplified expression of inflammatory genes, which is associated with increased acetylation of glucocorticoid receptors. Neutrophilic inflammation contributes to severe asthma and may be related to T helper (Th) 17 rather than Th2 cytokines. IL-17A levels are elevated in severe asthma and correlate with the presence of neutrophils. Restoring the activity of HDAC2 or targeting the Th17 signaling pathway is a potential therapeutic approach to reverse steroid insensitivity.
Collapse
Affiliation(s)
- Lihuan Ouyang
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Guomei Su
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Jingyun Quan
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Zhilin Xiong
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Tianwen Lai
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| |
Collapse
|
23
|
Abstract
Increasing evidence suggests that there is acceleration of lung ageing in chronic lung diseases, such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), with the accumulation of senescent cells in the lung. Senescent cells fail to repair tissue damage and release an array of inflammatory proteins, known as the senescence-associated secretory phenotype, which drive further senescence and disease progression. This suggests that targeting cellular senescence with senotherapies may treat the underlying disease process in COPD and IPF and thus reduce disease progression and mortality. Several existing or future drugs may inhibit the development of cellular senescence which is driven by chronic oxidative stress (senostatics), including inhibitors of PI3K-mTOR signalling pathways, antagomirs of critical microRNAs and novel antioxidants. Other drugs (senolytics) selectively remove senescent cells by promoting apoptosis. Clinical studies with senotherapies are already underway in chronic lung diseases.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart & Lung Institute, Imperial College London, United Kingdom.
| |
Collapse
|
24
|
Tiao-Bu-Fei-Shen Formula Improves Glucocorticoid Resistance of Chronic Obstructive Pulmonary Disease via Downregulating the PI3K-Akt Signaling Pathway and Promoting GR α Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2023; 2023:4359616. [PMID: 36820399 PMCID: PMC9938767 DOI: 10.1155/2023/4359616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/21/2022] [Accepted: 11/24/2022] [Indexed: 02/12/2023]
Abstract
Objective To predict and determine the mechanism through which Tiao-Bu-Fei-Shen (TBFS) formula improves glucocorticoid resistance in chronic obstructive pulmonary disease (COPD), using network pharmacology, molecular docking technology, and in vitro studies. Methods The main active components and associated targets of TBFS were screened using the systems pharmacology database of traditional Chinese medicine database (TCMSP). The main COPD targets were retrieved from the Human Gene (GeneCards) and DrugBank databases. A protein-protein interaction (PPI) network was constructed using the protein interaction platform STRING and Cytoscape 3.6.1. Gene ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genome Pathway (KEGG) analyses were performed using the biological information annotation database Metascape. Molecular docking was performed using the AutoDock Vina software. THP-1 monocytes were treated with TBFS-containing serum and cigarette smoke extract (CSE) for 48 h, and cell proliferation in each group was determined using cell counting kit-8 (CCK-8). A COPD cell model was constructed by stimulating THP-1 monocytes with CSE for 12 h. A lentivirus vector for RNA interference of histone deacetylase 2 (HDAC2) gene was constructed and transfected into the THP-1 monocytes, and the transfection efficiency was verified using quantitative polymerase chain reaction (qPCR) and western blotting (WB). The expression of HDAC2 in each group of cells was detected using qPCR, and the expression of HDAC2, phosphoinositide-3 kinase (PI3K) p85α, glucocorticoid receptor α (GRα), and P-AKT1 in each group of cells was detected through WB. Results A total of 344 TBFS active components, 249 related drug targets, 1,171 COPD target proteins, and 138 drug and disease intersection targets were obtained. Visual analysis of the PPI network map revealed that the core COPD targets of TBFS were AKT1, IL-6, TNF, TP53, and IL1-β. KEGG pathway enrichment analysis resulted in the identification of 20 signaling pathways as the main pathways involved in the action of TBFS against COPD, including the PI3K-Akt, TNF, and IL-17 signaling pathways. Molecular docking experiments revealed a strong binding capacity of kaempferol, luteolin, and quercetin to the ATK1 protein in TBFS, with quercetin performing the best. PCR results showed that treatment with TBFS significantly increased the expression levels of HDAC2 in the COPD model. WB results showed that TBFS treatment significantly increased the expression levels of GRα and HDAC2 in the COPD model, while reducing the expression levels of P-AKT1. Conclusion TBFS treatment improves glucocorticoid resistance observed in COPD through downregulation of the PI3K-Akt signaling pathway and promotion of GRα expression.
Collapse
|
25
|
Nishimoto Y, Kimura G, Ito K, Kizawa Y. [Anti-inflammatory Effects of a Src Inhibitor on the Murine Model of Asthma Exacerbation Induced by Ovalbumin and Lipopolysaccharide]. YAKUGAKU ZASSHI 2023; 143:191-197. [PMID: 36724932 DOI: 10.1248/yakushi.22-00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Asthma is often exacerbated by airway infection, and some patients with severe asthma may be unresponsive to conventional corticosteroid treatment. Src family kinases (SFKs) were recently implicated in the inflammatory responses of mice induced by allergen and bacterial toxin lipopolysaccharide (LPS). Therefore, we examined the effects of dasatinib (DAS), a Src inhibitor, on airway inflammation in mice induced by ovalbumin (OVA) and LPS. Male A/J mice were sensitized to OVA Day -14 and -7, challenged with intranasal OVA on Day 0, 2, 4, 6 and 8, and on Day 10, mice were also challenged with OVA via inhalation. Mice were treated intranasally with DAS or fluticasone propionate (FP), a glucocorticoid, twice daily for 3 d starting 1 d after OVA inhalation. Moreover, some mice were also administrated LPS 2 h after DAS or FP treatment to model of asthma exacerbation. One day after the last intervention, lung tissue and bronchoalveolar lavage fluid (BALF) were collected. DAS attenuated the accumulation of inflammatory cells and cytokines/chemokines in BALF induced by both OVA and OVA+LPS, while FP did not reduce accumulations induced by OVA+LPS. Therefore, targeting SFKs may be a superior therapeutic approach for asthma exacerbation by infection.
Collapse
Affiliation(s)
- Yuki Nishimoto
- Laboratory of Physiology and Anatomy, School of pharmacy, Nihon University
| | - Genki Kimura
- Laboratory of Physiology and Anatomy, School of pharmacy, Nihon University
| | - Kazuhiro Ito
- National Heart and Lung Institute, Imperial College London
| | - Yasuo Kizawa
- Laboratory of Physiology and Anatomy, School of pharmacy, Nihon University
| |
Collapse
|
26
|
Airway Smooth Muscle Regulated by Oxidative Stress in COPD. Antioxidants (Basel) 2023; 12:antiox12010142. [PMID: 36671004 PMCID: PMC9854973 DOI: 10.3390/antiox12010142] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
Since COPD is a heterogeneous disease, a specific anti-inflammatory therapy for this disease has not been established yet. Oxidative stress is recognized as a major predisposing factor to COPD related inflammatory responses, resulting in pathological features of small airway fibrosis and emphysema. However, little is known about effects of oxidative stress on airway smooth muscle. Cigarette smoke increases intracellular Ca2+ concentration and enhances response to muscarinic agonists in human airway smooth muscle. Cigarette smoke also enhances proliferation of these cells with altered mitochondrial protein. Hydrogen peroxide and 8-isoprostans are increased in the exhaled breath condensate in COPD. These endogenous oxidants cause contraction of tracheal smooth muscle with Ca2+ dynamics through Ca2+ channels and with Ca2+ sensitization through Rho-kinase. TNF-α and growth factors potentiate proliferation of these cells by synthesis of ROS. Oxidative stress can alter the function of airway smooth muscle through Ca2+ signaling. These phenotype changes are associated with manifestations (dyspnea, wheezing) and pathophysiology (airflow limitation, airway remodeling, airway hyperresponsiveness). Therefore, airway smooth muscle is a therapeutic target against COPD; oxidative stress should be included in treatable traits for COPD to advance precision medicine. Research into Ca2+ signaling related to ROS may contribute to the development of a novel agent for COPD.
Collapse
|
27
|
Xie T, Huang R, Deng D, Tang P, Fu Y, Zheng Y, Wan Y. Cryptotanshinone Reverses Corticosteroid Insensitivity by Inhibition of Phosphoinositide-3-Kinase-δ in Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2023; 18:797-809. [PMID: 37180749 PMCID: PMC10171224 DOI: 10.2147/copd.s405757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/30/2023] [Indexed: 05/16/2023] Open
Abstract
Purpose Corticosteroid insensitivity has become a major barrier in the treatment of chronic obstructive pulmonary disease (COPD). It is known that oxidative stress reduces the expression and activity of histone deacetylase (HDAC)-2 by activating phosphoinositide-3-kinase-δ(PI3Kδ)/Akt pathway, which is a common mechanism. The aim of this study was to investigate whether cryptotanshinone (CPT) can improve corticosteroid sensitivity and to investigate the molecular mechanisms by which this occurs. Patients and Methods Corticosteroid sensitivity in peripheral blood mononuclear cells (PBMCs) collected from COPD patients, or in human monocytic U937 monocytic cells exposed to cigarette smoke extract (CSE), was quantified as the dexamethasone concentration required to achieve 30% inhibition of tumor necrosis factor-α (TNFα)-induced interleukin 8 (IL-8) production in the presence or absence of cryptotanshinone. PI3K/Akt activity (measured as the relative ratio of phosphorylated Akt at Ser-473 to total Akt) and HDAC2 expression levels were determined by western blotting. HDAC activity was evaluated by a Fluo-Lys HDAC activity assay kit in U937 monocytic cells. Results Both PBMCs in patients with COPD and U937 cells exposed to CSE were found to be insensitive to dexamethasone, accompanied by increased phosphorylated Akt (pAkt) and decreased HDAC2 protein expression. The pretreatment of cryptotanshinone restored their sensitivity to dexamethasone, and simultaneously downregulated the level of phosphorylated Akt and upregulated the level of HDAC2 protein. Pretreatment with cryptotanshinone or IC87114 reversed the decrease in HDAC activity in CSE-stimulated U937 cells. Conclusion Cryptotanshinone restores corticosteroid sensitivity induced by oxidative stress via inhibition of PI3Kδ and is a potential treatment for corticosteroid-insensitive diseases such as COPD.
Collapse
Affiliation(s)
- Tao Xie
- Department of Respiratory Diseases, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Rong Huang
- Department of Respiratory Diseases, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Daishuo Deng
- Department of Respiratory Diseases, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| | - Peipei Tang
- Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai’an, Jiangsu, People’s Republic of China
| | - Yufeng Fu
- Institute of Medicinal Biotechnology, Jiangsu College of Nursing, Huai’an, Jiangsu, People’s Republic of China
| | - Yulong Zheng
- Department of Respiratory Diseases, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
- Correspondence: Yulong Zheng; Yufeng Wan, Department of Respiratory Diseases, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China, Tel +86 137 7670 7363; +86 158 0523 0282, Fax +86 517 8087 1636; +86 517 8087 1616, Email ;
| | - Yufeng Wan
- Department of Respiratory Diseases, The Affiliated Huai’an Hospital of Xuzhou Medical University, Huai’an, Jiangsu, People’s Republic of China
| |
Collapse
|
28
|
Naciff JM, Shan YK, Wang X, Daston GP. Article title: Transcriptional profiling efficacy to define biological activity similarity for cosmetic ingredients' safety assessment based on next-generation read-across. FRONTIERS IN TOXICOLOGY 2022; 4:1082222. [PMID: 36618549 PMCID: PMC9811170 DOI: 10.3389/ftox.2022.1082222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
The objective of this work was to use transcriptional profiling to assess the biological activity of structurally related chemicals to define their biological similarity and with that, substantiate the validity of a read-across approach usable in risk assessment. Two case studies are presented, one with 4 short alkyl chain parabens: methyl (MP), ethyl (EP), butyl (BP), and propylparaben (PP), as well as their main metabolite, p-hydroxybenzoic acid (pHBA) with the assumption that propylparaben was the target chemical; and a second one with caffeine and its main metabolites theophylline, theobromine and paraxanthine where CA was the target chemical. The comprehensive transcriptional response of MCF7, HepG2, A549 and ICell cardiomyocytes was evaluated (TempO-Seq) after exposure to vehicle-control, each paraben or pHBA, CA or its metabolites, at 3 non-cytotoxic concentrations, for 6 h. Differentially expressed genes (FDR ≥0.05, and fold change ±1.2≥) were identified for each chemical, at each concentration, and used to determine similarities. Each of the chemicals is able to elicit changes in the expression of a number of genes, as compared to controls. Importantly, the transcriptional profile elicited by each of the parabens shares a high degree of similarity across the group. The highest number of genes commonly affected was between butylparaben and PP. The transcriptional profile of the parabens is similar to the one elicited by estrogen receptor agonists, with BP being the closest structural and biological analogue for PP. In the CA case, the transcriptional profile elicited of all four methylxanthines had a high degree of similarity across the cell types, with CA and theophylline being the most active. The most robust response was obtained in the cardiomyocytes with the highest transcriptional profile similarity between CA and TP. The transcriptional profile of the methylxanthines is similar to the one elicited by inhibitors of phosphatidylinositol 3-kinase as well as other kinase inhibitors. Overall, our results support the approach of incorporating transcriptional profiling in well-designed in vitro tests as one robust stream of data to support biological similarity driven read-across procedures and strengthening the traditional structure-based approaches useful in risk assessment.
Collapse
|
29
|
Zhang R, Liu H, Dai D, Ding X, Wang D, Wang Y, Shi X, Zhang S, Duan X, Wang H, Luo Y, Liu S, Han B, Zhang X, Fang Y, Yang J, Xu W, Sun T. Adjunctive sepsis therapy with aminophylline (STAP): a randomized controlled trial. Chin Med J (Engl) 2022; 135:2843-2850. [PMID: 36728571 PMCID: PMC9944697 DOI: 10.1097/cm9.0000000000002282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Sepsis is a serious disease caused by infection. Aminophylline has anti-asthma and anti-inflammatory effects. We aimed to explore the safety and effect of aminophylline in sepsis. METHODS We conducted a clinical randomized controlled trial involving 100 patients diagnosed with sepsis within 48 h after intensive care unit (ICU) admission in two sites. All patients were randomized in a 1:1 ratio to receive standard therapy with or without aminophylline. The primary clinical outcome was all-cause mortality at 28 days. RESULTS From September 27, 2018 to February 12, 2020, we screened 277 septic patients and eventually enrolled 100 patients, with 50 assigned to the aminophylline group and 50 to the usual-care group. At 28 days, 7 of 50 patients (14.0%) in the aminophylline group had died, compared with 16 of 50 (32.0%) in the usual-care group ( P = 0.032). Cox regression showed that the aminophylline group had a lower hazard of death (hazard ratio = 0.312, 95% confidence interval: 0.129-0.753). Compared with the usual-care group, patients in the aminophylline group had a longer survival time ( P = 0.039 by the log-rank test). The effects of aminophylline on vasopressor dose, oxygenation index, and sequential organ failure assessment score were time-dependent with treatment. There were no significant differences in total hospitalization days, ICU hospitalization days, and rates of serious adverse events (all P > 0.05). No adverse events were observed in the trial. CONCLUSIONS Aminophylline as an adjunct therapy could significantly reduce the risk of death and prolong the survival time of patients with sepsis. TRIAL REGISTRATION ChiCTR.org.cn, ChiCTR1800019173.
Collapse
Affiliation(s)
- Ruifang Zhang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Huan Liu
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Dongmei Dai
- Department of Intensive Care Unit, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Xianfei Ding
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Dong Wang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Yan Wang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Xuexiu Shi
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Shuguang Zhang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Xiaoguang Duan
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Haixu Wang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Yonggang Luo
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Shaohua Liu
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Bing Han
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Xiaojuan Zhang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Yu Fang
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| | - Jing Yang
- Precision Medicine Monitoring Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Wangbin Xu
- Department of Intensive Care Unit, the First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Tongwen Sun
- General ICU, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou, Henan 450052, China
| |
Collapse
|
30
|
Elekhnawy E, Negm WA, El-Sherbeni SA, Zayed A. Assessment of drugs administered in the Middle East as part of the COVID-19 management protocols. Inflammopharmacology 2022; 30:1935-1954. [PMID: 36018432 PMCID: PMC9411846 DOI: 10.1007/s10787-022-01050-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/03/2022] [Indexed: 02/06/2023]
Abstract
The pandemic spread of coronavirus (COVID-19) has been reported first at the end of 2019. It continues disturbing various human aspects with multiple pandemic waves showing more fatal novel variants. Now Egypt faces the sixth wave of the pandemic with controlled governmental measures. COVID-19 is an infectious respiratory disease-causing mild to moderate illness that can be progressed into life-threatening complications based on patients- and variant type-related factors. The symptoms vary from dry cough, fever to difficulty in breathing that required urgent hospitalization. Most countries have authorized their national protocols for managing manifested symptoms and thus lowering the rate of patients' hospitalization and boosting the healthcare systems. These protocols are still in use even with the development and approval of several vaccines. These protocols were instructed to aid home isolation, bed rest, dietary supplements, and additionally the administration of antipyretic, steroids, and antiviral drugs. The current review aimed to highlight the administered protocols in the Middle East, namely in Egypt and the Kingdom of Saudi Arabia demonstrating how these protocols have shown potential effectiveness in treating patients and saving many soles.
Collapse
Affiliation(s)
- Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Elguish Street (Medical Campus), Tanta, 31527 Egypt
| | - Walaa A. Negm
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Elguish Street (Medical Campus), Tanta, 31527 Egypt
| | - Suzy A. El-Sherbeni
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Elguish Street (Medical Campus), Tanta, 31527 Egypt
| | - Ahmed Zayed
- Pharmacognosy Department, Faculty of Pharmacy, Tanta University, Elguish Street (Medical Campus), Tanta, 31527 Egypt
- Institute of Bioprocess Engineering, Technical University of Kaiserslautern, Gottlieb-Daimler-Straße 49, 67663 Kaiserslautern, Germany
| |
Collapse
|
31
|
Albano GD, Gagliardo RP, Montalbano AM, Profita M. Overview of the Mechanisms of Oxidative Stress: Impact in Inflammation of the Airway Diseases. Antioxidants (Basel) 2022; 11:2237. [PMID: 36421423 PMCID: PMC9687037 DOI: 10.3390/antiox11112237] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/09/2022] [Accepted: 11/11/2022] [Indexed: 08/01/2023] Open
Abstract
Inflammation of the human lung is mediated in response to different stimuli (e.g., physical, radioactive, infective, pro-allergenic or toxic) such as cigarette smoke and environmental pollutants. They often promote an increase in inflammatory activities in the airways that manifest themselves as chronic diseases (e.g., allergic airway diseases, asthma, chronic bronchitis/chronic obstructive pulmonary disease (COPD) or even lung cancer). Increased levels of oxidative stress (OS) reduce the antioxidant defenses, affect the autophagy/mitophagy processes, and the regulatory mechanisms of cell survival, promoting inflammation in the lung. In fact, OS potentiate the inflammatory activities in the lung, favoring the progression of chronic airway diseases. OS increases the production of reactive oxygen species (ROS), including superoxide anions (O2-), hydroxyl radicals (OH) and hydrogen peroxide (H2O2), by the transformation of oxygen through enzymatic and non-enzymatic reactions. In this manner, OS reduces endogenous antioxidant defenses in both nucleated and non-nucleated cells. The production of ROS in the lung can derive from both exogenous insults (cigarette smoke or environmental pollution) and endogenous sources such as cell injury and/or activated inflammatory and structural cells. In this review, we describe the most relevant knowledge concerning the functional interrelation between the mechanisms of OS and inflammation in airway diseases.
Collapse
|
32
|
Smoking Cessation in Mice Does Not Switch off Persistent Lung Inflammation and Does Not Restore the Expression of HDAC2 and SIRT1. Int J Mol Sci 2022; 23:ijms23169104. [PMID: 36012370 PMCID: PMC9409159 DOI: 10.3390/ijms23169104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022] Open
Abstract
Once COPD is established, pulmonary lesions can only progress and smoking cessation by itself is not sufficient to switch off persistent lung inflammation. Similarly, in former-smoker mice, neutrophil inflammation persists and lung lesions undergo progressive deterioration. The molecular mechanisms underlying disease progression and the inefficiency of smoking cessation in quenching neutrophilic inflammation were studied in male C57 Bl/6 mice after 6 months of rest from smoking cessation. As compared with the mice that continued to smoke, the former-smoker mice showed reduced expression of histone deacetylases HDAC2 and SIRT1 and marked expression of p-p38 MAPK and p-Ser10. All these factors are involved in corticosteroid insensitivity and in perpetuating inflammation. Former-smoker mice do show persistent lung neutrophilic influx and a high number of macrophages which account for the intense staining in the alveolar structures of neutrophil elastase and MMP-9 (capable of destroying lung scaffolding) and 8-OHdG (marker of oxidative stress). “Alarmins” released from necrotic cells together with these factors can sustain and perpetuate inflammation after smoking cessation. Several factors and mechanisms all together are involved in sustaining and perpetuating inflammation in former-smoker mice. This study suggests that a better control of COPD in humans may be achieved by precise targeting of the various molecular mechanisms associated with different phenotypes of disease by using a cocktail of drug active toward specific molecules.
Collapse
|
33
|
Barnes PJ. Oxidative Stress in Chronic Obstructive Pulmonary Disease. Antioxidants (Basel) 2022; 11:antiox11050965. [PMID: 35624831 PMCID: PMC9138026 DOI: 10.3390/antiox11050965] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 12/16/2022] Open
Abstract
There is a marked increase in oxidative stress in the lungs of patients with COPD, as measured by increased exhaled 8-isoprostane, ethane, and hydrogen peroxide in the breath. The lung may be exposed to exogenous oxidative stress from cigarette smoking and indoor or outdoor air pollution and to endogenous oxidative stress from reactive oxygen species released from activated inflammatory cells, particularly neutrophils and macrophages, in the lungs. Oxidative stress in COPD may be amplified by a reduction in endogenous antioxidants and poor intake of dietary antioxidants. Oxidative stress is a major driving mechanism of COPD through the induction of chronic inflammation, induction of cellular senescence and impaired autophagy, reduced DNA repair, increased autoimmunity, increased mucus secretion, and impaired anti-inflammatory response to corticosteroids. Oxidative stress, therefore, drives the pathology of COPD and may increase disease progression, amplify exacerbations, and increase comorbidities through systemic oxidative stress. This suggests that antioxidants may be effective as disease-modifying treatments. Unfortunately, thiol-based antioxidants, such as N-acetylcysteine, have been poorly effective, as they are inactivated by oxidative stress in the lungs, so there is a search for more effective and safer antioxidants. New antioxidants in development include mitochondria-targeted antioxidants, NOX inhibitors, and activators of the transcription factor Nrf2, which regulates several antioxidant genes.
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart and Lung Institute, Imperial College London, London SW5 9LH, UK
| |
Collapse
|
34
|
Montaño LM, Sommer B, Gomez-Verjan JC, Morales-Paoli GS, Ramírez-Salinas GL, Solís-Chagoyán H, Sanchez-Florentino ZA, Calixto E, Pérez-Figueroa GE, Carter R, Jaimez-Melgoza R, Romero-Martínez BS, Flores-Soto E. Theophylline: Old Drug in a New Light, Application in COVID-19 through Computational Studies. Int J Mol Sci 2022; 23:ijms23084167. [PMID: 35456985 PMCID: PMC9030606 DOI: 10.3390/ijms23084167] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023] Open
Abstract
Theophylline (3-methyxanthine) is a historically prominent drug used to treat respiratory diseases, alone or in combination with other drugs. The rapid onset of the COVID-19 pandemic urged the development of effective pharmacological treatments to directly attack the development of new variants of the SARS-CoV-2 virus and possess a therapeutical battery of compounds that could improve the current management of the disease worldwide. In this context, theophylline, through bronchodilatory, immunomodulatory, and potentially antiviral mechanisms, is an interesting proposal as an adjuvant in the treatment of COVID-19 patients. Nevertheless, it is essential to understand how this compound could behave against such a disease, not only at a pharmacodynamic but also at a pharmacokinetic level. In this sense, the quickest approach in drug discovery is through different computational methods, either from network pharmacology or from quantitative systems pharmacology approaches. In the present review, we explore the possibility of using theophylline in the treatment of COVID-19 patients since it seems to be a relevant candidate by aiming at several immunological targets involved in the pathophysiology of the disease. Theophylline down-regulates the inflammatory processes activated by SARS-CoV-2 through various mechanisms, and herein, they are discussed by reviewing computational simulation studies and their different applications and effects.
Collapse
Affiliation(s)
- Luis M. Montaño
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, CP, Mexico; (L.M.M.); (R.J.-M.); (B.S.R.-M.)
| | - Bettina Sommer
- Laboratorio de Hiperreactividad Bronquial, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Ciudad de México 14080, CP, Mexico;
| | - Juan C. Gomez-Verjan
- Dirección de Investigación, Instituto Nacional de Geriatría, Ciudad de México 10200, CP, Mexico; (J.C.G.-V.); (G.S.M.-P.)
| | - Genaro S. Morales-Paoli
- Dirección de Investigación, Instituto Nacional de Geriatría, Ciudad de México 10200, CP, Mexico; (J.C.G.-V.); (G.S.M.-P.)
| | - Gema Lizbeth Ramírez-Salinas
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica (Laboratory for the Design and Development of New Drugs and Biotechnological Innovation), Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón S/N, Col. Santo Tomas, Ciudad de México 11340, CP, Mexico;
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, Ciudad de México 14510, CP, Mexico
| | - Héctor Solís-Chagoyán
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Ciudad de México 14370, CP, Mexico; (H.S.-C.); (Z.A.S.-F.)
| | - Zuly A. Sanchez-Florentino
- Laboratorio de Neurofarmacología, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Ciudad de México 14370, CP, Mexico; (H.S.-C.); (Z.A.S.-F.)
| | - Eduardo Calixto
- Departamento de Neurobiología, Dirección de Investigación en Neurociencias, Instituto Nacional de Psiquiatría “Ramón de la Fuente Muñiz”, Ciudad de México 14370, CP, Mexico;
| | - Gloria E. Pérez-Figueroa
- Instituto Nacional de Neurología y Neurocirugía, Unidad Periférica en el Estudio de la Neuroinflamación en Patologías Neurológicas, Ciudad de México 06720, CP, Mexico;
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Ciudad de México 06720, CP, Mexico
| | - Rohan Carter
- FRACGP/MBBS, Murchison Outreach Service Mount Magnet Western Australia, Mount Magnet, WA 6530, Australia;
| | - Ruth Jaimez-Melgoza
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, CP, Mexico; (L.M.M.); (R.J.-M.); (B.S.R.-M.)
| | - Bianca S. Romero-Martínez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, CP, Mexico; (L.M.M.); (R.J.-M.); (B.S.R.-M.)
| | - Edgar Flores-Soto
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, CP, Mexico; (L.M.M.); (R.J.-M.); (B.S.R.-M.)
- Correspondence: ; Tel.: +52-555-6232279
| |
Collapse
|
35
|
The Use of Inhaled Corticosteroids for Patients with COPD Who Continue to Smoke Cigarettes: An Evaluation of Current Practice. Am J Med 2022; 135:302-312. [PMID: 34655541 DOI: 10.1016/j.amjmed.2021.09.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 11/22/2022]
Abstract
The use of inhaled corticosteroids (ICS) in combination with inhaled bronchodilators for patients with chronic obstructive pulmonary disease (COPD) is a common practice in primary care settings. However, ICS-containing therapies may be less effective in patients with COPD compared with asthma, and in individuals with COPD who continue to smoke cigarettes. Preclinical studies suggest that inflammation in COPD is very different from in asthma. Glucocorticoid receptor functioning and other innate anti-inflammatory mechanisms are altered in cells exposed to cigarette smoke. COPD may be relatively insensitive to ICS, especially in individuals who continue to smoke. ICS-containing therapies in patients with asthma who continue to smoke may also be less effective compared with patients who do not smoke. ICS-containing therapies may be inappropriately used in some patients with COPD, and their long-term use is associated with an increased risk for side effects, including pneumonia and bone fractures in some patients. Treatment for patients with COPD should be carefully evaluated, and anti-inflammatory/bronchodilatory strategies should be chosen based on individual patient characteristics and recommendations in current guidelines.
Collapse
|
36
|
Chen X, Wang D, Guo X, Li X, Ye W, Qi Y, Gu W. Curcumin-Loaded mPEG-PLGA Nanoparticles Attenuates the Apoptosis and Corticosteroid Resistance Induced by Cigarette Smoke Extract. Front Pharmacol 2022; 13:824652. [PMID: 35281918 PMCID: PMC8914114 DOI: 10.3389/fphar.2022.824652] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/07/2022] [Indexed: 01/14/2023] Open
Abstract
The present study was aim to prepare curcumin-loaded methoxypolyethylene-glycols-poly (lactic-co-glycolic acid) nanoparticles (Cur-mPEG-PLGA-NPs) and investigate curcumin’s effect on reversing corticosteroid resistance induced by cigarette smoke extract (CSE) in rat tracheal epithelial (RTE) cells. The Cur-mPEG-PLGA-NPs were spherical, regular in shape with smooth surfaces, and well distributed and Cur-mPEG-PLGA-NP suspensions had good water solubility and presented prolonged release. Furthermore, we found that Cur-mPEG-PLGA-NPs were internalized more than curcumin into the cells and significantly alleviated apoptosis in RTE cells. In addition, 10% CSE reduced the maximal inhibition percentage and increased the half-inhibitory concentration of budesonide (BUD) on IL-8 secretion, and curcumin restored the efficacy of BUD inhibition. BUD in combination with Cur-mPEG-PLGA-NPs showed higher inhibitory rates for LPS- and CSE-induced IL-8 secretion than that in combination with curcumin. Moverover, the relative expression levels of HDAC2 was reduced after CSE exposure and curcumin could improve HDAC2 expression and reverse CSE-induced corticosteroid resistance. Curcumin in high concentration and Cur-mPEG-PLGA-NPs restored HDAC2 levels in RTE cells and thus Cur-mPEG-PCL-NPs have higher biological activity than curcumin.
Collapse
Affiliation(s)
- Xi Chen
- Department of Respirology Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Di Wang
- Department of Intensive Care Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xuejun Guo
- Department of Respirology Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaoming Li
- Department of Respirology Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Wenjing Ye
- Department of Respirology Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yang Qi
- Department of Pharmacy, No. 900 Hospital of the Chinese PLA Joint Support Forces, Fuzhou, China
- *Correspondence: Yang Qi, ; Wen Gu,
| | - Wen Gu
- Department of Respirology Medicine, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- *Correspondence: Yang Qi, ; Wen Gu,
| |
Collapse
|
37
|
Abstract
The characteristic features of chronic obstructive pulmonary disease (COPD) include inflammation and remodelling of the lower airways and lung parenchyma together with activation of inflammatory and immune processes. Due to the increasing habit of cigarette smoking worldwide COPD prevalence is increasing globally. Current therapies are unable to prevent COPD progression in many patients or target many of its hallmark characteristics which may reflect the lack of adequate biomarkers to detect the heterogeneous clinical and molecular nature of COPD. In this chapter we review recent molecular data that may indicate novel pathways that underpin COPD subphenotypes and indicate potential improvements in the classes of drugs currently used to treat COPD. We also highlight the evidence for new drugs or approaches to treat COPD identified using molecular and other approaches including kinase inhibitors, cytokine- and chemokine-directed biologicals and small molecules, antioxidants and redox signalling pathway inhibitors, inhaled anti-infectious agents and senolytics. It is important to consider the phenotypes/molecular endotypes of COPD patients together with specific outcome measures to target new therapies to particular COPD subtypes. This will require greater understanding of COPD molecular pathologies and a focus on biomarkers of predicting disease subsets and responder/non-responder populations.
Collapse
Affiliation(s)
- Isabel Uwagboe
- Airways Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| | - Ian M Adcock
- Airways Disease Section, National Heart and Lung Institute, Imperial College, London, UK -
| | - Federica Lo Bello
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università di Messina, Messina, Italy
| | - Sharon Mumby
- Airways Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| |
Collapse
|
38
|
To Y. [Drug-repositioning indicates a way to conquest corticosteroid-insensitivity]. Nihon Yakurigaku Zasshi 2022; 157:286-292. [PMID: 36047137 DOI: 10.1254/fpj.22021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Recent advance of medications and devices brings more effective treatment intervention to the patients with asthma. As far as obeying guidelines, approximately 90% of patients with asthma acquire good control. However, there are still small number of patients with asthma who resist conventional treatment. Most of them are corticosteroid-insensitive. It would be thought that there are two ways to deal with this problem. The first one is biologics. However, these are very expensive. The second way is a treatment intervention focusing on cancellation of corticosteroid-resistance. In early 2010s, a new-intracellular signaling pathway (phosphatydilinositol-3-kinese: PI3K pathway) is proven to be closely associated with corticosteroid-resistance. PI3K-inhibitor should be one of the promising candidates to attenuate corticosteroid-resistance. But PI3K-inhibitor also has a toxicity when given systemically. Drug-repositioning (DR) is a good option to deal with it. To find out PI3K-inhibitor from numerous medicines gives an answer how to inhibit PI3K pathway. The presenter has found both low-dose theophylline and long-acting beta-2 agonist have a potential to antagonize PI3K. These medicines have been prescribed for decades and their safety has already been proved. It is possible for clinicians to inhibit PI3K activation in patients with asthma associated with corticosteroid-resistance using these medicines without waiting for development of bran-new PI3K inhibitors. DR revealed that nortriptyline acts as a steroid-enhancer via its anti-PI3K activity. Calcium-channel blocker saves lung function in patients with asthma in long-term observation. DR promises us to find cheap and safe options to treat difficult asthma by inhibiting specific intracellular signaling pathways.
Collapse
Affiliation(s)
- Yasuo To
- Department of Pulmonary Medicine, International University of Health and Welfare School of Medicine
- Department of Pulmonary Medicine, International University of Health and Welfare Atami Hospital
| |
Collapse
|
39
|
Novel Immunomodulatory Therapies for Respiratory Pathologies. COMPREHENSIVE PHARMACOLOGY 2022. [PMCID: PMC8238403 DOI: 10.1016/b978-0-12-820472-6.00073-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
40
|
Fagone E, Fruciano M, Gili E, Sambataro G, Vancheri C. Developing PI3K Inhibitors for Respiratory Diseases. Curr Top Microbiol Immunol 2022; 436:437-466. [DOI: 10.1007/978-3-031-06566-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
41
|
|
42
|
Faniyi AA, Hughes MJ, Scott A, Belchamber KBR, Sapey E. Inflammation, Ageing and Diseases of the Lung: Potential therapeutic strategies from shared biological pathways. Br J Pharmacol 2021; 179:1790-1807. [PMID: 34826882 DOI: 10.1111/bph.15759] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/07/2021] [Accepted: 11/08/2021] [Indexed: 12/15/2022] Open
Abstract
Lung diseases disproportionately affect elderly individuals. The lungs form a unique environment: a highly elastic organ with gaseous exchange requiring the closest proximity of inhaled air containing harmful agents and the circulating blood volume. The lungs are highly susceptible to senescence, with age and "inflammageing" creating a pro-inflammatory environment with a reduced capacity to deal with challenges. Whilst lung diseases may have disparate causes, the burden of ageing and inflammation provides a common process which can exacerbate seemingly unrelated pathologies. However, these shared pathways may also provide a common route to treatment, with increased interest in drugs which target ageing processes across respiratory diseases. In this review, we will examine the evidence for the increased burden of lung disease in older adults, the structural and functional changes seen with advancing age and assess what our expanding knowledge of inflammation and ageing pathways could mean for the treatment of lung disease.
Collapse
Affiliation(s)
- A A Faniyi
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| | - M J Hughes
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| | - A Scott
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| | - K B R Belchamber
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| | - E Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, U.K
| |
Collapse
|
43
|
Kadushkin AG, Tahanovich AD, Movchan LV, Kolesnikova TS, Khadasouskaya EV, Shman TV. The Effect of Glucocorticoids in Combination with Azithromycin or Theophylline on Cytokine Production by NK and NKT-Like Blood Cells of Patients with Chronic Obstructive Pulmonary Disease. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES B: BIOMEDICAL CHEMISTRY 2021. [DOI: 10.1134/s1990750821040053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
44
|
Audousset C, McGovern T, Martin JG. Role of Nrf2 in Disease: Novel Molecular Mechanisms and Therapeutic Approaches - Pulmonary Disease/Asthma. Front Physiol 2021; 12:727806. [PMID: 34658913 PMCID: PMC8511424 DOI: 10.3389/fphys.2021.727806] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/02/2021] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a major transcription factor involved in redox homeostasis and in the response induced by oxidative injury. Nrf2 is present in an inactive state in the cytoplasm of cells. Its activation by internal or external stimuli, such as infections or pollution, leads to the transcription of more than 500 elements through its binding to the antioxidant response element. The lungs are particularly susceptible to factors that generate oxidative stress such as infections, allergens and hyperoxia. Nrf2 has a crucial protective role against these ROS. Oxidative stress and subsequent activation of Nrf2 have been demonstrated in many human respiratory diseases affecting the airways, including asthma and chronic obstructive pulmonary disease (COPD), or the pulmonary parenchyma such as acute respiratory distress syndrome (ARDS) and pulmonary fibrosis. Several compounds, both naturally occurring and synthetic, have been identified as Nrf2 inducers and enhance the activation of Nrf2 and expression of Nrf2-dependent genes. These inducers have proven particularly effective at reducing the severity of the oxidative stress-driven lung injury in various animal models. In humans, these compounds offer promise as potential therapeutic strategies for the management of respiratory pathologies associated with oxidative stress but there is thus far little evidence of efficacy through human trials. The purpose of this review is to summarize the involvement of Nrf2 and its inducers in ARDS, COPD, asthma and lung fibrosis in both human and in experimental models.
Collapse
Affiliation(s)
- Camille Audousset
- Meakins-Christie Laboratories, McGill University, Montréal, QC, Canada
| | - Toby McGovern
- Meakins-Christie Laboratories, McGill University, Montréal, QC, Canada
| | - James G Martin
- Meakins-Christie Laboratories, McGill University, Montréal, QC, Canada
| |
Collapse
|
45
|
Dobric A, De Luca SN, Spencer SJ, Bozinovski S, Saling MM, McDonald CF, Vlahos R. Novel pharmacological strategies to treat cognitive dysfunction in chronic obstructive pulmonary disease. Pharmacol Ther 2021; 233:108017. [PMID: 34626675 DOI: 10.1016/j.pharmthera.2021.108017] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/19/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major incurable global health burden and currently the 3rd largest cause of death in the world, with approximately 3.23 million deaths per year. Globally, the financial burden of COPD is approximately €82 billion per year and causes substantial morbidity and mortality. Importantly, much of the disease burden and health care utilisation in COPD is associated with the management of its comorbidities and viral and bacterial-induced acute exacerbations (AECOPD). Recent clinical studies have shown that cognitive dysfunction is present in up to 60% of people with COPD, with impairments in executive function, memory, and attention, impacting on important outcomes such as quality of life, hospitalisation and survival. The high prevalence of cognitive dysfunction in COPD may also help explain the insufficient adherence to therapeutic plans and strategies, thus worsening disease progression in people with COPD. However, the mechanisms underlying the impaired neuropathology and cognition in COPD remain largely unknown. In this review, we propose that the observed pulmonary oxidative burden and inflammatory response of people with COPD 'spills over' into the systemic circulation, resulting in damage to the brain and leading to cognitive dysfunction. As such, drugs targeting the lungs and comorbidities concurrently represent an exciting and unique therapeutic opportunity to treat COPD and cognitive impairments, which may lead to the production of novel targets to prevent and reverse the debilitating and life-threatening effects of cognitive dysfunction in COPD.
Collapse
Affiliation(s)
- Aleksandar Dobric
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Simone N De Luca
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Sarah J Spencer
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia; ARC Centre of Excellence for Nanoscale Biophotonics, RMIT University, Melbourne, VIC, Australia
| | - Steven Bozinovski
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Michael M Saling
- Clinical Neuropsychology, The University of Melbourne and Austin Health, VIC, Australia
| | - Christine F McDonald
- Institute for Breathing and Sleep, Austin Health, Melbourne, VIC, Australia; Department of Respiratory & Sleep Medicine, The University of Melbourne and Austin Health, Melbourne, VIC, Australia
| | - Ross Vlahos
- School of Health & Biomedical Sciences, RMIT University, Melbourne, VIC, Australia.
| |
Collapse
|
46
|
Redox Regulation in Aging Lungs and Therapeutic Implications of Antioxidants in COPD. Antioxidants (Basel) 2021; 10:antiox10091429. [PMID: 34573061 PMCID: PMC8470212 DOI: 10.3390/antiox10091429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 08/27/2021] [Accepted: 09/01/2021] [Indexed: 12/23/2022] Open
Abstract
Mammals, including humans, are aerobic organisms with a mature respiratory system to intake oxygen as a vital source of cellular energy. Despite the essentiality of reactive oxygen species (ROS) as byproducts of aerobic metabolism for cellular homeostasis, excessive ROS contribute to the development of a wide spectrum of pathological conditions, including chronic lung diseases such as COPD. In particular, epithelial cells in the respiratory system are directly exposed to and challenged by exogenous ROS, including ozone and cigarette smoke, which results in detrimental oxidative stress in the lungs. In addition, the dysfunction of redox regulation due to cellular aging accelerates COPD pathogenesis, such as inflammation, protease anti-protease imbalance and cellular apoptosis. Therefore, various drugs targeting oxidative stress-associated pathways, such as thioredoxin and N-acetylcysteine, have been developed for COPD treatment to precisely regulate the redox system. In this review, we present the current understanding of the roles of redox regulation in the respiratory system and COPD pathogenesis. We address the insufficiency of current COPD treatment as antioxidants and discuss future directions in COPD therapeutics targeting oxidative stress while avoiding side effects such as tumorigenesis.
Collapse
|
47
|
Kadushkin AG, Tahanovich AD, Movchan LV, Kolesnikova TS, Khadasouskaya AV, Shman TV. [The effect of glucocorticoids in combination with azithromycin or theophylline on cytokine production by NK and NKT-like blood cells of patients with chronic obstructive pulmonary disease]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2021; 67:352-359. [PMID: 34414894 DOI: 10.18097/pbmc20216704352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by reduced sensitivity of cells to the anti-inflammatory effects of glucocorticoids (GCs). Azithromycin and a low dose theophylline have a significant impact on molecular mechanisms leading to corticosteroid resistance. The aim of this study was to evaluate the ability of azithromycin and theophylline to enhance the anti-inflammatory effects of GCs on the production of cytokines by NK and NKT-like blood cells of COPD patients. Whole blood cells from COPD patients (n=21) were incubated in the presence of budesonide (10 nM), azithromycin (10 μg/mL), theophylline (1 μM), or their combinations and stimulated with phorbol myristate acetate (50 ng/mL). Intracellular production of proinflammatory cytokines in NK (CD3-CD56+) and NKT-like (CD3+CD56+) blood cells was analyzed by flow cytometry. Budesonide reduced synthesis of interleukin 4 (IL-4), CXCL8, tumor necrosis factor α (TNFα) by NK and NKT-like cells, as well as production of interferon γ (IFNγ) by NK cells. Azithromycin suppressed production of IL-4 and CXCL8 by NK and NKT-like cells, and theophylline inhibited IL-4 synthesis by these lymphocytes. The combination of azithromycin and budesonide had a more pronounced inhibitory effect on the production of IL-4 and CXCL8 by NK and NKT-like cells than the effect of these drugs alone. The combination of theophylline and budesonide suppressed synthesis of IL-4 and CXCL8 by NK and NKT-like cells, as well as the production of TNFα and IFNγ by NK cells stronger than budesonide alone. The obtained results demonstrate the benefits for the combined use of GCs with theophylline at a low dose or azithromycin to suppress the inflammatory process in patients with COPD.
Collapse
Affiliation(s)
| | | | - L V Movchan
- Republican Scientific and Practical Center of Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | | | | | - T V Shman
- Republican Scientific and Practical Center of Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| |
Collapse
|
48
|
Role of Histone Deacetylases in Monocyte Function in Health and Chronic Inflammatory Diseases. Rev Physiol Biochem Pharmacol 2021; 180:1-47. [PMID: 33974124 DOI: 10.1007/112_2021_59] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Histone deacetylases (HDACs) are a family of 18 members that participate in the epigenetic regulation of gene expression. In addition to histones, some HDACs also deacetylate transcription factors and specific cytoplasmic proteins.Monocytes, as part of the innate immune system, maintain tissue homeostasis and help fight infections and cancer. In these cells, HDACs are involved in multiple processes including proliferation, migration, differentiation, inflammatory response, infections, and tumorigenesis. Here, a systematic description of the role that most HDACs play in these functions is reviewed. Specifically, some HDACs induce a pro-inflammatory response and play major roles in host defense. Conversely, other HDACs reprogram monocytes and macrophages towards an immunosuppressive phenotype. The right balance between both types helps monocytes to respond correctly to the different physiological/pathological stimuli. However, aberrant expressions or activities of specific HDACs are associated with autoimmune diseases along with other chronic inflammatory diseases, infections, or cancer.This paper critically reviews the interesting and extensive knowledge regarding the role of some HDACs in these pathologies. It also shows that as yet, very little progress has been made toward the goal of finding effective HDAC-targeted therapies. However, given their obvious potential, we conclude that it is worth the effort to develop monocyte-specific drugs that selectively target HDAC subtypes with the aim of finding effective treatments for diseases in which our innate immune system is involved.
Collapse
|
49
|
Zheng X, Chen D, Zhu X, Le Grange JM, Zhou L, Zhang J. Impacts of anti-inflammatory phosphodiesterase inhibitors on a murine model of chronic pulmonary inflammation. Pharmacol Res Perspect 2021; 9:e00840. [PMID: 34327862 PMCID: PMC8322673 DOI: 10.1002/prp2.840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 01/04/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) often tends to respond poorly to glucocorticoid (GC) therapy. Reduced Histone deacetylase-2 (HDAC-2) activity is an important mechanism behind this GC insensitivity. In this study, we investigated the effects of three phosphodiesterase inhibitors (PDEIs), with an anti-inflammatory propensity, on cigarette smoke (CS)-induced pulmonary inflammation and HDAC-2 activity. Male C57BL/6 mice were exposed to cigarette smoke (CS) over the course of 30 weeks. Administration of the PDEIs commenced from the 29th week and followed a schedule of once daily treatments, 5 days a week, for 2 weeks. Roflumilast (ROF) was administered intragastrically (5 mg·kg-1 ), while pentoxifylline (PTX) (10 mg·kg-1 ) and theophylline (THEO) (10 mg·kg-1 ) were administered intraperitoneally, either alone or in combination with a GC (triamcinolone acetonide or TRI, 5 mg·kg-1 , i.m., single injection). Lung morphometry, as well as the activity of HDAC-2, pro-inflammatory cytokines and reactive oxygen species (ROS) were assessed at the end of the 30-week course. CS exposure was associated with a reduction in HDAC-2 activity and the up-regulation of ROS expression. PTX, ROF, and THEO administration led to the partial restoration of HDAC-2 activity, which was favorably associated with the reduction of ROS expression. However, combining TRI to any of these PDEIs did not synergistically augment HDAC-2 activity. Inactivation of HDAC-2 due to long-term CS exposure is closely related to exaggerated oxidative stress, and this reduced HDAC-2 activity could partially be restored through the use of PDEIs. This finding provides a potential novel approach for further clinical research.
Collapse
Affiliation(s)
- Xiao‐Fang Zheng
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Dan‐Dan Chen
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiao‐Ling Zhu
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jehane Michael Le Grange
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lu‐Qian Zhou
- Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Jin‐Nong Zhang
- Department of Emergency MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
50
|
Liu G, Särén L, Douglasson H, Zhou XH, Åberg PM, Ollerstam A, Betts CJ, Balogh Sivars K. Precision cut lung slices: an ex vivo model for assessing the impact of immunomodulatory therapeutics on lung immune responses. Arch Toxicol 2021; 95:2871-2877. [PMID: 34191076 DOI: 10.1007/s00204-021-03096-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 06/15/2021] [Indexed: 11/28/2022]
Abstract
Chronic inflammatory diseases of the respiratory tract, such as chronic obstructive pulmonary disease (COPD) and asthma, are severe lung diseases that require effective treatments. In search for new medicines for these diseases, there is an unmet need for predictive and translatable disease-relevant in vitro/ex vivo models to determine the safety and efficacy of novel drug candidates. Here, we report the use of precision cut lung slices (PCLS) as a potential ex vivo platform to study compound effects in a physiologically relevant environment. PCLS derived from an elastase-challenged mouse model display key characteristics of increased inflammation ex vivo, which is exacerbated further upon challenge with LPS, mimicking the immune insult of a pathogen triggering disease exacerbation. Such LPS-induced inflammatory conditions are significantly abrogated by immunomodulatory agents targeting specific inflammatory signaling pathways in the absence of cytotoxic effects in lung slices. Thus, an ex vivo model of PCLS with a simulated pathogenic insult can replicate proposed in vivo pharmacological effects and thus could potentially act as a valuable tool to investigate the underlying mechanisms associated with lung safety, therapeutic efficacy and exacerbations with infection.
Collapse
Affiliation(s)
- Guanghui Liu
- Respiratory and Immunology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Linnea Särén
- Animal Sciences and Technologies, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Helena Douglasson
- Bioscience Cough and in Vivo, Early Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xiao-Hong Zhou
- Patient Safety, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Per M Åberg
- Respiratory and Immunology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Ollerstam
- Respiratory and Immunology Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Catherine J Betts
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
| | - Kinga Balogh Sivars
- Clinical Testing, Global Procurement, Operations, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|