1
|
Xie D, Quan J, Yu X, Liang Z, Chen Y, Wu L, Lin L, Fan L. Molecular mechanism of Jianpiyifei II granules in the treatment of chronic obstructive pulmonary disease: Network pharmacology analysis, molecular docking, and experimental assessment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155273. [PMID: 38342020 DOI: 10.1016/j.phymed.2023.155273] [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: 02/06/2023] [Revised: 10/24/2023] [Accepted: 12/10/2023] [Indexed: 02/13/2024]
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is defined by persistent airway and lung inflammation, excessive mucus production, remodeling of the airways, and damage to the alveolar tissue. Based on clinical experience, it has been observed that Jianpiyifei II (JPYF II) granules exhibit a significant therapeutic impact on individuals suffering from stable COPD. Nevertheless, the complete understanding of JPYF II's potential mode of action against COPD remains to be further clarified. PURPOSE To further investigate the underlying mechanism of JPYF II for treating COPD and clarify the role of the IL-17 pathway in the treatment. METHODS A variety of databases were utilized to acquire JPYF II's bioactive components, as well as related targets of JPYF II and COPD. Cytoscape was utilized to establish multiple interaction networks for the purpose of topological analyses and core-target screening. The Metascape was utilized to identify the function of target genes and crucial signaling pathways. To evaluate the interactions between bioactive ingredients and central target proteins, molecular docking simulations were conducted. Following that, a sequence of experiments was conducted both in the laboratory and in living organisms, which included analyzing the cell counts in bronchoalveolar lavage fluid (BALF), examining lung tissue for histopathological changes, conducting immunohistochemistry, RT‒qPCR, ELISA, and Western blotting. RESULTS In JPYF II, 88 bioactive ingredients were predicted to have a total of 342 targets. After conducting Venn analysis, it was discovered that 284 potential targets of JPYF II were linked to the provision of defensive benefits against COPD. The PPI network yielded a total of twenty-four core targets. The findings from the analysis of enrichment and gene‒pathway network suggested that JPYF II targeted Hsp90, MAPKs, ERK, AP-1, TNF-α, IL-6, COX-2, CXCL8, and MMP-9 as crucial elements for COPD treatment through the IL-17 pathway. Additionally, JPYF II might modulate MAPK signaling pathways and the downstream transcription factor AP-1 via IL-17 regulation. According to the findings from molecular docking, it was observed that the 24 core target proteins exhibited robust binding affinities towards the top 10 bioactive compounds. Furthermore, the treatment of COPD through the regulation of MAPKs in the IL-17 pathway was significantly influenced by flavonoids and sterols found in JPYF II. In vitro, these observations were further confirmed. In vivo results demonstrated that JPYF II reduced inflammatory cell infiltration in pulmonary tissues and the quantity of inflammatory cells in BALF obtained from LPS- and CS-stimulated mice. Moreover, the administration of JPYF II resulted in the inhibition of IL-17 mRNA and protein levels, phosphorylation levels of MAPK proteins, and expression of phosphorylated AP-1 proteins. It also suppressed the expression of downstream effector genes and proteins associated with the IL-17/MAPK/AP-1 signaling axis in lung tissues and BALF. CONCLUSION This research reveals that JPYF II improves COPD by controlling the IL-17/MAPK/AP-1 signaling axis within the IL-17 pathway for the first time. These findings offer potential approaches for the creation of novel medications that specifically target IL-17 and proteins involved in the IL-17 pathway to address COPD.
Collapse
Affiliation(s)
- Dan Xie
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; Guangdong‒Hong Kong‒Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Jingyu Quan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; Guangdong‒Hong Kong‒Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xuhua Yu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; Guangdong‒Hong Kong‒Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Ziyao Liang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; Guangdong‒Hong Kong‒Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Yuanbin Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; Guangdong‒Hong Kong‒Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Lei Wu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; Guangdong‒Hong Kong‒Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China.
| | - Lin Lin
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; Guangdong‒Hong Kong‒Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China.
| | - Long Fan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China; Guangdong‒Hong Kong‒Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China.
| |
Collapse
|
2
|
Henen C, Johnson EA, Wiesel S. Unleashing the Power of IL-17: A Promising Frontier in Chronic Obstructive Pulmonary Disease (COPD) Treatment. Cureus 2023; 15:e41977. [PMID: 37465090 PMCID: PMC10350480 DOI: 10.7759/cureus.41977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2023] [Indexed: 07/20/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a pulmonary ailment that is both degenerative and incapacitating, with a global prevalence affecting millions. Despite notable progress in treatment methodologies, there is still a critical requirement for innovative therapeutic interventions. The pathogenesis of COPD has recently seen a significant focus on the role of interleukin 17 (IL-17), a pro-inflammatory cytokine. This review investigates the potential of IL-17 targeting as a viable therapeutic approach for treating COPD. The literature indicates a complex correlation between IL-17 and COPD. Research has indicated that IL-17 plays a role in the manifestation of airway inflammation, remodeling, and mucus hypersecretion, considered characteristic attributes of COPD. Elevated levels of IL-17 have been observed in the lungs of individuals with COPD, indicating its potential as a therapeutic target for intervention. Furthermore, preclinical studies utilizing animal models of COPD have demonstrated the efficacy of anti-IL-17 antibodies in reducing airway inflammation and remodeling. Comprehending the mechanical principles that underlie IL-17 signaling in COPD is imperative for advancing focused, therapeutic interventions. Activating diverse signaling pathways, such as the β-catenin and Act 1 adaptor protein (ACT 1) mediated pathways, is a crucial aspect of COPD pathogenesis triggered by IL-17. As a result, the suppression of IL-17 signaling has exhibited encouraging outcomes in mitigating pulmonary hypertension induced by hypoxia and interrupting the signaling mediated by ACT 1. Notwithstanding these promising discoveries, additional investigation is required to comprehensively explain the function of IL-17 in COPD and its viability as a target for therapy. The efficacy and safety of biological treatments that target IL-17 in COPD patients necessitate thorough investigation despite their initial positive outcomes. Furthermore, identifying appropriate patient subpopulations that would benefit most from IL-17-targeted therapies and optimizing treatment protocols are binding domains for future investigation. The current review presents a persuasive case for the imperative requirement of an additional investigation into the targeting of IL-17 for COPD management. Through a comprehensive analysis of the complex relationship between IL-17 and COPD pathogenesis, novel therapeutic avenues can be explored, potentially transforming the approach to managing this incapacitating condition. As we explore this novel domain, the possibility of pioneering therapies aimed at IL-17 presents a ray of optimism for the multitudes of individuals afflicted with the onerous consequences of COPD.
Collapse
Affiliation(s)
- Christine Henen
- Internal Medicine, St. John's Episcopal Hospital, New York, USA
| | - Elise A Johnson
- Medical School, Ross University School of Medicine, Miramar, USA
| | - Shimshon Wiesel
- Pulmonary/Critical Care/Sleep Medicine, St. John's Episcopal Hospital, New York, USA
| |
Collapse
|
3
|
Pei Y, Wei Y, Peng B, Wang M, Xu W, Chen Z, Ke X, Rong L. Combining single-cell RNA sequencing of peripheral blood mononuclear cells and exosomal transcriptome to reveal the cellular and genetic profiles in COPD. Respir Res 2022; 23:260. [PMID: 36127695 PMCID: PMC9490964 DOI: 10.1186/s12931-022-02182-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/09/2022] [Indexed: 11/30/2022] Open
Abstract
Background It has been a long-held consensus that immune reactions primarily mediate the pathology of chronic obstructive pulmonary disease (COPD), and that exosomes may participate in immune regulation in COPD. However, the relationship between exosomes and peripheral immune status in patients with COPD remains unclear. Methods In this study, we sequenced plasma exosomes and performed single-cell RNA sequencing on peripheral blood mononuclear cells (PBMCs) from patients with COPD and healthy controls. Finally, we constructed competing endogenous RNA (ceRNA) and protein–protein interaction (PPI) networks to delineate the interactions between PBMCs and exosomes within COPD. Results We identified 135 mRNAs, 132 lncRNAs, and 359 circRNAs from exosomes that were differentially expressed in six patients with COPD compared with four healthy controls. Functional enrichment analyses revealed that many of these differentially expressed RNAs were involved in immune responses including defending viral infection and cytokine–cytokine receptor interaction. We also identified 18 distinct cell clusters of PBMCs in one patient and one control by using an unsupervised cluster analysis called uniform manifold approximation and projection (UMAP). According to resultant cell identification, it was likely that the proportions of monocytes, dendritic cells, and natural killer cells increased in the COPD patient we tested, meanwhile the proportions of B cells, CD4 + T cells, and naïve CD8 + T cells declined. Notably, CD8 + T effector memory CD45RA + (Temra) cell and CD8 + effector memory T (Tem) cell levels were elevated in patient with COPD, which were marked by their lower capacity to differentiate due to their terminal differentiation state and lower reactive capacity to viral pathogens. Conclusions We generated exosomal RNA profiling and single-cell transcriptomic profiling of PBMCs in COPD, described possible connection between impaired immune function and COPD development, and finally determined the possible role of exosomes in mediating local and systemic immune reactions. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02182-8.
Collapse
Affiliation(s)
- Yanli Pei
- Respiratory Medicine Department, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yuxi Wei
- Peking Union Medical College (PUMC), PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Boshizhang Peng
- Peking Union Medical College (PUMC), PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Mengqi Wang
- Peking Union Medical College (PUMC), PUMC and Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Xu
- Respiratory Medicine Department, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhe Chen
- Laboratory of Cough, Affiliated Kunshan Hospital of Jiangsu University, Suzhou, Jiangsu, China.
| | - Xindi Ke
- Peking Union Medical College (PUMC), PUMC and Chinese Academy of Medical Sciences, Beijing, China.
| | - Lei Rong
- Respiratory Medicine Department, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
4
|
Chitosan-Coated Solid Lipid Nano-Encapsulation Improves the Therapeutic Antiairway Inflammation Effect of Berberine against COPD in Cigarette Smoke-Exposed Rats. Can Respir J 2022; 2022:8509396. [PMID: 35465190 PMCID: PMC9033382 DOI: 10.1155/2022/8509396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/29/2022] [Indexed: 02/08/2023] Open
Abstract
Berberine (Ber) is an isoquinoline alkaloid that has shown therapeutic potential in mice with chronic obstructive pulmonary disease (COPD). However, the therapeutic efficiency of Ber is restricted by its low aqueous solubility and bioavailability. Chitosan and solid lipid nanoparticles (SLNs) have demonstrated great abilities as delivery systems in enhancing the bioavailability of therapeutic compounds. The present study aimed to get together the biological features of SLNs with the advantages of chitosan to formulate an efficient nano-carrier platform for the oral delivery of Ber and evaluate the therapeutic effect of the prepared Ber-encapsulated nanoparticles on airway inflammation in cigarette smoke (CS)-induced COPD rats. The Ber-encapsulated SLE-chitosan formulation was manufactured using a modified solvent-injection method followed by a homogenization process. Physicochemical properties, encapsulation efficiency, in vitro stability and Ber release, and pharmacokinetics of the manufactured formulation were evaluated. The COPD rat model was developed by exposing animals to CS. To study the therapeutic efficiency of Ber-encapsulated SLE-chitosan nanoparticles and pure berberine, the histopathological changes of the lung tissues, levels of inflammatory cells and cytokines, and activities of myeloperoxidase (MPO) and superoxide dismutase (SOD) enzymes were evaluated in bronchoalveolar lavage fluid (BALF). Ber-encapsulated SLE-chitosan showed the particle size in nano-range with high stability and controlled slow-release profile in vitro in simulated gastric (pH 1.5) and intestinal (pH 6.8) fluids. Administration of Ber-loaded SLE-chitosan nanoparticles could significantly ameliorate inflammation scores in lung tissues and reduce levels of inflammatory cells (neutrophils and macrophages) and inflammatory cytokines (IL-1β, Il-6, Il-17, and TNFα) in BALF when compared with the pure Ber. SLE-chitosan-based nanoparticles can strongly improve the therapeutic anti-inflammatory impact of Ber against CS-induced airway inflammation in COPD rats, suggesting the promising application of Ber-encapsulated SLN-chitosan nanoparticles for treating COPD and other inflammation-mediated diseases.
Collapse
|
5
|
Ouyang S, Liu C, Xiao J, Chen X, Lui AC, Li X. Targeting IL-17A/glucocorticoid synergy to CSF3 expression in neutrophilic airway diseases. JCI Insight 2020; 5:132836. [PMID: 32051346 DOI: 10.1172/jci.insight.132836] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 01/08/2020] [Indexed: 12/15/2022] Open
Abstract
IL-17A plays a critical role in the pathogenesis of steroid-resistant neutrophilic airway inflammation, which is a hallmark of severe asthma and chronic obstructive pulmonary disease (COPD). Through RNA sequencing analysis of transcriptomes of human airway smooth muscle cells treated with IL-17A, dexamethasone (DEX, a synthetic glucocorticoid drug), alone or in combination, we identified a group of genes that are synergistically induced by IL-17A and DEX, including the neutrophil-promoting cytokine CSF3. In type-17 (Th17/IL-17Ahi) preclinical models of neutrophilic severe asthma (acute and chronic) and COPD, although DEX treatment was able to reduce the expression of neutrophil-mobilizing CXCL1 and CXCL2 in lung tissue, CSF3 expression was upregulated by DEX treatment. We found that DEX treatment alone failed to alleviate neutrophilic airway inflammation and pathology, and even exacerbated the disease phenotype when CSF3 was highly induced. Disruption of the IL-17A/DEX synergy by IL-17A inhibition with anti-IL-17A mAb or cyanidin-3-glucoside (C3G, a small-molecule IL-17A blocker) or depletion of CSF3 effectively rendered DEX sensitivity in type-17 preclinical models of neutrophilic airway diseases. Our study elucidates what we believe is a novel mechanism of steroid resistance in type-17 neutrophilic airway inflammation and offers an effective steroid-sparing therapeutic strategy (combined low-dose DEX and C3G) for treating neutrophilic airway diseases.
Collapse
|
6
|
Xiong J, Tian J, Zhou L, Le Y, Sun Y. Interleukin-17A Deficiency Attenuated Emphysema and Bone Loss in Mice Exposed to Cigarette Smoke. Int J Chron Obstruct Pulmon Dis 2020; 15:301-310. [PMID: 32103929 PMCID: PMC7020917 DOI: 10.2147/copd.s235384] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/22/2020] [Indexed: 12/20/2022] Open
Abstract
Background and Purpose Chronic obstructive pulmonary disease (COPD) is a common chronic inflammatory disease, which is associated with various comorbidities including osteoporosis. Interleukin(IL)-17 has been reported to play important roles in the pathogenesis of COPD and also associated with bone destruction in inflammatory diseases. However, the role of IL-17A in COPD-related osteoporosis is yet unknown. The purpose of our study was to investigate the potential contribution of IL-17A in COPD-related bone loss. Materials and Methods We examined the bone mass and bone microarchitecture in wild-type and IL-17A-/- mice exposed to long-term cigarette smoke (CS). Osteoclast activities and the expression of receptor activator of nuclear factor-κB ligand (RANKL) in bone tissues were assessed, and the blood levels of inflammatory cytokines were measured. Results Less bone loss as well as attenuated emphysema were shown in IL-17A-/- mice compared with wild-type mice. CS-exposed IL-17A-/- mice had decreased TRAP+ osteoclast numbers and lower RANKL expression compared with CS-exposed wild-type mice. Inflammatory cytokines including IL-6 and IL-1β in circulation were decreased in IL-17A-/- mice exposed to CS compared with wild-type mice. Conclusion This study indicates that IL-17A is involved in CS-induced bone loss and may be a common link between COPD and osteoporosis.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Jieyu Tian
- Hematology Oncology Center, Beijing Children's Hospital, Capital Medical University, Beijing 100045, People's Republic of China
| | - Lu Zhou
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Yanqing Le
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Yongchang Sun
- Department of Respiratory and Critical Care Medicine, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| |
Collapse
|
7
|
Hersh CP, Zacharia S, Prakash Arivu Chelvan R, Hayden LP, Mirtar A, Zarei S, Putcha N. Immunoglobulin E as a Biomarker for the Overlap of Atopic Asthma and Chronic Obstructive Pulmonary Disease. CHRONIC OBSTRUCTIVE PULMONARY DISEASES-JOURNAL OF THE COPD FOUNDATION 2020; 7:1-12. [PMID: 31999898 DOI: 10.15326/jcopdf.7.1.2019.0138] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Asthma-COPD overlap (ACO) is a common clinical syndrome, yet there is no single objective definition. We hypothesized that immunoglobulin E (IgE) measurements could be used to refine the definition of ACO. In baseline plasma samples from 2870 participants in the COPD Genetic Epidemiology (COPDGene®) study, we measured total IgE levels and specific IgE levels to 6 common allergens. Compared to usual chronic obstructive pulmonary disease (COPD), participants with ACO (based on self-report of asthma) had higher total IgE levels (median 67.0 versus 42.2 IU/ml) and more frequently had at least one positive specific IgE (43.5% versus 24.5%). We previously used a strict definition of ACO in participants with COPD, based on self-report of a doctor's diagnosis of asthma before age 40. This strict ACO definition was refined by the presence of atopy, determined by total IgE > 100 IU/ml or at least one positive specific IgE, as was the broader definition of ACO based on self-reported asthma history. Participants with all 3 ACO definitions were younger (mean age 60.0-61.3 years), were more commonly African American (36.8%-44.2%), had a higher exacerbation frequency (1.0-1.2 in the past year), and had more airway wall thickening on quantitative analysis of chest computed tomography (CT) scans. Among participants with ACO, 37%-46% did not have atopy; these individuals had more emphysema on chest CT scan. Based on associations with exacerbations and CT airway disease, IgE did not clearly improve the clinical definition of ACO. However, IgE measurements could be used to subdivide individuals with atopic and non-atopic ACO, who might have different biologic mechanisms and potential treatments.
Collapse
Affiliation(s)
- Craig P Hersh
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Soumya Zacharia
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Lystra P Hayden
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Sara Zarei
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts.,Arasila Biotech, San Diego, California
| | - Nirupama Putcha
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland
| | | |
Collapse
|
8
|
Zhang XW, Liu W, Jiang HL, Mao B. Dissection of Pharmacological Mechanism of Chinese Herbal Medicine Yihuo Huatan Formula on Chronic Obstructive Pulmonary Disease: A Systems Pharmacology-Based Study. Sci Rep 2019; 9:13431. [PMID: 31530860 PMCID: PMC6748909 DOI: 10.1038/s41598-019-50064-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 09/04/2019] [Indexed: 02/05/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the most common respiratory diseases. Yihuo Huatan Formula (YHF), as a proven Chinese Herbal Medicine (CHM), has been verified to be effective in the treatment of stable COPD through years' of practice. Nevertheless, its working mechanism is still unclear. We sought to systematically decipher the mechanism of YHF for treating stable COPD using systems pharmacology-based method that integrates pharmacokinetic screening, target prediction, network analyses, GO and KEGG enrichment analyses. Firstly, a total of 1267 chemicals out of 15 herbal components were included in YHF chemical database. Among them, 180 potential active molecules were screened out through pharmacokinetic evaluation. Then 258 targets of the active molecules were predicted, of which 84 were chosen for further analyses. Finally, the network analyses and GO and KEGG enrichment methods suggested a therapeutic effect of YHF on the alleviation of airway inflammation, decrease of mucus secretion, maintenance of immune homeostasis and benefit of COPD comorbidities, by regulating multiple targets and pathways. The systems pharmacology-based approach helps to understand the underlying working mechanism of YHF in stable COPD from a holistic perspective, and offers an exemplification for systematically uncovering the action mechanisms of CHM.
Collapse
Affiliation(s)
- Xia-Wei Zhang
- Division of Respiratory Medicine, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan, 610041, P. R. China
| | - Wei Liu
- Division of Respiratory Medicine, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan, 610041, P. R. China
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy, West China Hospital, West China School of Medicine, Sichuan University, 1 Keyuansi Road, Chengdu, Sichuan, 610041, P. R. China
| | - Hong-Li Jiang
- Division of Respiratory Medicine, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan, 610041, P. R. China.
| | - Bing Mao
- Division of Respiratory Medicine, Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, 37 Guoxue Lane, Chengdu, Sichuan, 610041, P. R. China
| |
Collapse
|
9
|
Ke Q, Yang L, Cui Q, Diao W, Zhang Y, Xu M, He B. Ciprofibrate attenuates airway remodeling in cigarette smoke-exposed rats. Respir Physiol Neurobiol 2019; 271:103290. [PMID: 31525465 DOI: 10.1016/j.resp.2019.103290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023]
Abstract
Airway remodeling is a key pathological lesion in chronic obstructive pulmonary disease (COPD), and it leads to poorly reversible airway obstruction. Current pharmacological interventions are ineffective at controlling airway remodeling. To address this issue, we queried the Connectivity Map (cMap) database to screen for drug candidates that had the potential to dilate the bronchus and inhibit airway smooth muscle (ASM) proliferation. We identified ciprofibrate as a drug candidate. Ciprofibrate inhibited cigarette smoke extract-induced rat ASM cell contraction and proliferation in vitro. We exposed Sprague-Dawley (SD) rats to clean air or cigarette smoke (CS) and treated the rats with ciprofibrate. Ciprofibrate improved pulmonary function, inhibited airway hypercontraction, and ameliorated morphological small airway remodeling, including airway smooth muscle proliferation, in CS-exposed rats. Ciprofibrate also significantly reduced IL-1β, IL-12p70, IL-17A and IL-18 expression, which are related to airway remodeling, in the sera of CS-exposed rats. These findings indicate that ciprofibrate could attenuate airway remodeling in CS-exposed rats.
Collapse
Affiliation(s)
- Qian Ke
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China.
| | - Lin Yang
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China.
| | - Qinghua Cui
- Department of Biomedical Informatics, Department of Physiology and Pathophysiology, Center for Noncoding RNA Medicine, MOE Key Lab of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Wenqi Diao
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China.
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China; Beijing Key Laboratory of Cardiovascular Receptors Research. Beijing, China.
| | - Ming Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China; NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China; Beijing Key Laboratory of Cardiovascular Receptors Research. Beijing, China; Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Bei He
- Department of Respiratory Medicine, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
10
|
Hersh CP. Pharmacogenomics of chronic obstructive pulmonary disease. Expert Rev Respir Med 2019; 13:459-470. [PMID: 30925849 PMCID: PMC6482089 DOI: 10.1080/17476348.2019.1601559] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/27/2019] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Chronic obstructive pulmonary disease (COPD) is a heterogeneous condition, which presents the opportunity for precision therapy based on genetics or other biomarkers. Areas covered: Alpha-1 antitrypsin deficiency, a genetic form of emphysema, provides an example of this precision approach to diagnosis and therapy. To date, research in COPD pharmacogenomics has been limited by small sample sizes, lack of accessible target tissue, failure to consider COPD subtypes, and different outcomes relevant for various medications. There have been several published genome-wide association studies and other omics studies in COPD pharmacogenomics; however, clinical implementation remains far away. There is a growing evidence base for precision prescription of inhaled corticosteroids in COPD, based on clinical phenotypes and blood biomarkers, but not yet based on pharmacogenomics. Expert opinion: At this time, there is insufficient evidence for clinical implementation of COPD pharmacogenomics. Additional genome-wide studies will be required to discover predictors of drug response and to identify genomic biomarkers of COPD subtypes, which could be targeted with subtype-directed therapies.
Collapse
Affiliation(s)
- Craig P Hersh
- a Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine , Brigham and Women's Hospital, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
11
|
Chakraborti S, Sarkar J, Chakraborti T. Role of PLD-PKCζ signaling axis in p47phox phosphorylation for activation of NADPH oxidase by angiotensin II in pulmonary artery smooth muscle cells. Cell Biol Int 2019; 43:678-694. [PMID: 30977575 DOI: 10.1002/cbin.11145] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 04/07/2019] [Indexed: 12/12/2022]
Abstract
We sought to determine the mechanism by which angiotensin II (ANGII) stimulates NADPH oxidase-mediated superoxide (O2 .- ) production in bovine pulmonary artery smooth muscle cells (BPASMCs). ANGII-induced increase in phospholipase D (PLD) and NADPH oxidase activities were inhibited upon pretreatment of the cells with chemical and genetic inhibitors of PLD2, but not PLD1. Immunoblot study revealed that ANGII treatment of the cells markedly increases protein kinase C-α (PKC-α), -δ, -ε, and -ζ levels in the cell membrane. Pretreatment of the cells with chemical and genetic inhibitors of PKC-ζ, but not PKC-α, -δ, and -ε, attenuated ANGII-induced increase in NADPH oxidase activity without a discernible change in PLD activity. Transfection of the cells with p47phox small interfering RNA inhibited ANGII-induced increase in NADPH oxidase activity without a significant change in PLD activity. Pretreatment of the cells with the chemical and genetic inhibitors of PLD2 and PKC-ζ inhibited ANGII-induced p47phox phosphorylation and subsequently translocation from cytosol to the cell membrane, and also inhibited its association with p22phox (a component of membrane-associated NADPH oxidase). Overall, PLD-PKCζ-p47phox signaling axis plays a crucial role in ANGII-induced increase in NADPH oxidase-mediated O2 .- production in the cells.
Collapse
Affiliation(s)
- Sajal Chakraborti
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal 741235, India
| | - Jaganmay Sarkar
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal 741235, India
| | - Tapati Chakraborti
- Department of Biochemistry and Biophysics, University of Kalyani, Kalyani, West Bengal 741235, India
| |
Collapse
|
12
|
Chakraborty A, Boer JC, Selomulya C, Plebanski M, Royce SG. Insights into endotoxin-mediated lung inflammation and future treatment strategies. Expert Rev Respir Med 2018; 12:941-955. [PMID: 30221563 DOI: 10.1080/17476348.2018.1523009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Airway inflammatory disorders are prevalent diseases in need of better management and new therapeutics. Immunotherapies offer a solution to the problem of corticosteroid resistance. Areas covered: The current review focuses on lipopolysaccharide (Gram-negative bacterial endotoxin)-mediated inflammation in the lung and the animal models used to study related diseases. Endotoxin-induced lung pathology is usually initiated by antigen presenting cells (APC). We will discuss different subsets of APC including lung dendritic cells and macrophages, and their role in responding to endotoxin and environmental challenges. Expert commentary: The pharmacotherapeutic considerations to combat airway inflammation should cost-effectively improve quality of life with sustainable and safe strategies. Selectively targeting APCs in the lung offer the potential for a promising new strategy for the better management and treatment of inflammatory lung disease.
Collapse
Affiliation(s)
- Amlan Chakraborty
- a Department of Chemical Engineering , Monash University , Clayton , Australia.,b Department of Immunology and Pathology , Central Clinical School, Monash University , Melbourne , Australia
| | - Jennifer C Boer
- b Department of Immunology and Pathology , Central Clinical School, Monash University , Melbourne , Australia
| | - Cordelia Selomulya
- a Department of Chemical Engineering , Monash University , Clayton , Australia
| | - Magdalena Plebanski
- b Department of Immunology and Pathology , Central Clinical School, Monash University , Melbourne , Australia.,c School of Health and Biomedical Sciences and Enabling Capability platforms, Biomedical and Health Innovation , RMIT University , Melbourne , Australia
| | - Simon G Royce
- d Central Clinical School , Monash University , Clayton , Victoria , Australia.,e Department of Pharmacology , Monash University , Clayton , Australia
| |
Collapse
|
13
|
George T, Chakraborty M, Giembycz MA, Newton R. A bronchoprotective role for Rgs2 in a murine model of lipopolysaccharide-induced airways inflammation. Allergy Asthma Clin Immunol 2018; 14:40. [PMID: 30305828 PMCID: PMC6166284 DOI: 10.1186/s13223-018-0266-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/22/2018] [Indexed: 02/07/2023] Open
Abstract
Background Asthma exacerbations are associated with the recruitment of neutrophils to the lungs. These cells release proteases and mediators, many of which act at G protein-coupled receptors (GPCRs) that couple via Gq to promote bronchoconstriction and inflammation. Common asthma therapeutics up-regulate expression of the regulator of G protein signalling (RGS), RGS2. As RGS2 reduces signaling from Gq-coupled GPCRs, we have defined role(s) for this GTPase-activating protein in an acute neutrophilic model of lung inflammation. Methods Wild type and Rgs2−/− C57Bl6 mice were exposed to nebulized lipopolysaccharide (LPS). Lung function (respiratory system resistance and compliance) was measured using a SCIREQ flexivent small animal ventilator. Lung inflammation was assessed by histochemistry, cell counting and by cytokine and chemokine expression in bronchoalveolar lavage (BAL) fluid. Results Lipopolysaccharide inhalation induced transient airways hyperreactivity (AHR) and neutrophilic lung inflammation. While AHR and inflammation was greatest 3 h post-LPS exposure, BAL neutrophils persisted for 24 h. At 3 h post-LPS inhalation, multiple inflammatory cytokines (CSF2, CSF3, IL6, TNF) and chemokines (CCL3, CCL4, CXCL1, CXCL2) were highly expressed in the BAL fluid, prior to declining by 24 h. Compared to wild type counterparts, Rgs2−/− mice developed significantly greater airflow resistance in response to inhaled methacholine (MCh) at 3 h post-LPS exposure. At 24 h post-LPS exposure, when lung function was recovering in the wild type animals, MCh-induced resistance was increased, and compliance decreased, in Rgs2−/− mice. Thus, Rgs2−/− mice show AHR and stiffer lungs 24 h post-LPS exposure. Histological markers of inflammation, total and differential cell counts, and major cytokine and chemokine expression in BAL fluid were similar between wild type and Rgs2−/− mice. However, 3 and 24 h post-LPS exposure, IL12B expression was significantly elevated in BAL fluid from Rgs2−/− mice compared to wild type animals. Conclusions While Rgs2 is bronchoprotective in acute neutrophilic inflammation, no clear anti-inflammatory effect was apparent. Nevertheless, elevated IL12B expression in Rgs2−/− animals raises the possibility that RGS2 could dampen Th1 responses. These findings indicate that up-regulation of RGS2, as occurs in response to inhaled corticosteroids and long-acting β2-adrenoceptor agonists, may be beneficial in acute neutrophilic exacerbations of airway disease, including asthma. Electronic supplementary material The online version of this article (10.1186/s13223-018-0266-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tresa George
- 1Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| | - Mainak Chakraborty
- 2Immunology Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| | - Mark A Giembycz
- 1Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| | - Robert Newton
- 1Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4Z6 Canada
| |
Collapse
|
14
|
Hogmalm A, Bry M, Bry K. Pulmonary IL-1β expression in early life causes permanent changes in lung structure and function in adulthood. Am J Physiol Lung Cell Mol Physiol 2018; 314:L936-L945. [DOI: 10.1152/ajplung.00256.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Chorioamnionitis, mechanical ventilation, oxygen therapy, and postnatal infection promote inflammation in the newborn lung. The long-term consequences of pulmonary inflammation during infancy have not been well characterized. The aim of this study was to examine the impact of inflammation during the late saccular to alveolar stages of lung development on lung structure and function in adulthood. To induce IL-1β expression in the pulmonary epithelium of mice with a tetracycline-inducible human IL-1β transgene, doxycycline was administered via intraperitoneal injections to bitransgenic pups and their littermate controls on postnatal days (PN) 0, 0.5, and 1. Lung structure, inflammation, and airway reactivity were studied in adulthood. IL-1β production in early life resulted in increased numbers of macrophages and neutrophils on PN21, but inflammation subsided by PN42. Permanent changes in alveolar structure, i.e., larger alveoli and thicker alveolar walls, were present from PN21 to PN84. Lack of alveolar septation thus persisted after IL-1β production and inflammation had ceased. Early IL-1β production caused goblet cell hyperplasia, enhanced calcium-activated chloride channel 3 (CLCA3) protein expression, and increased airway reactivity in response to methacholine on PN42. Lymphoid follicles were present adjacent to small airways in the lungs of adult bitransgenic mice, and levels of the B cell chemoattractant CXC-motif ligand (CXCL) 13 were elevated in the lungs of bitransgenic mice compared with controls. In conclusion, IL-1β-induced pulmonary inflammation in early life causes a chronic lung disease in adulthood.
Collapse
Affiliation(s)
- Anna Hogmalm
- Department of Pediatrics, Institution of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maija Bry
- The Queen Silvia Children’s Hospital, Gothenburg, Sweden
| | - Kristina Bry
- Department of Pediatrics, Institution of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- The Queen Silvia Children’s Hospital, Gothenburg, Sweden
| |
Collapse
|