1
|
Patel VJ, Joharapurkar A, Jain MR. The Perspective of Using Flow Cytometry for Unpuzzling Hypoxia-Inducible Factors Signalling. Drug Res (Stuttg) 2024; 74:113-122. [PMID: 38350634 DOI: 10.1055/a-2248-9180] [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: 02/15/2024]
Abstract
Hypoxia-inducible factors (HIFs) are transcription factors that are responsible for adapting to the changes in oxygen levels in the cellular environment. HIF activity determines the expression of cellular proteins that control the development and physiology of the cells and pathophysiology of a disease. Understanding the role of specific HIF (HIF-1-3) in cellular function is essential for development of the HIF-targeted therapies. In this review, we have discussed the use of flow cytometry in analysing HIF function in cells. Proper understanding of HIF-signalling will help to design pharmacological interventions HIF-mediated therapy. We have discussed the role of HIF-signalling in various diseases such as cancer, renal and liver diseases, ulcerative colitis, arthritis, diabetes and diabetic complications, psoriasis, and wound healing. We have also discussed protocols that help to decipher the role of HIFs in these diseases that would eventually help to design promising therapies.
Collapse
Affiliation(s)
- Vishal J Patel
- Department of Pharmacology and Toxicology, Zydus Research Centre, Zydus Lifesciences Limited, Moraiya, Ahmedabad, India
| | - Amit Joharapurkar
- Department of Pharmacology and Toxicology, Zydus Research Centre, Zydus Lifesciences Limited, Moraiya, Ahmedabad, India
| | - Mukul R Jain
- Department of Pharmacology and Toxicology, Zydus Research Centre, Zydus Lifesciences Limited, Moraiya, Ahmedabad, India
| |
Collapse
|
2
|
Foresto-Neto O, da Silva ARPA, Cipelli M, Santana-Novelli FPR, Camara NOS. The impact of hypoxia-inducible factors in the pathogenesis of kidney diseases: a link through cell metabolism. Kidney Res Clin Pract 2023; 42:561-578. [PMID: 37448286 PMCID: PMC10565456 DOI: 10.23876/j.krcp.23.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/20/2023] [Accepted: 03/20/2023] [Indexed: 07/15/2023] Open
Abstract
Kidneys are sensitive to disturbances in oxygen homeostasis. Hypoxia and activation of the hypoxia-inducible factor (HIF) pathway alter the expression of genes involved in the metabolism of renal and immune cells, interfering with their functioning. Whether the transcriptional activity of HIF protects the kidneys or participates in the pathogenesis of renal diseases is unclear. Several studies have indicated that HIF signaling promotes fibrosis in experimental models of kidney disease. Other reports showed a protective effect of HIF activation on kidney inflammation and injury. In addition to the direct effect of HIF on the kidneys, experimental evidence indicates that HIF-mediated metabolic shift activates inflammatory cells, supporting the HIF cascade as a link between lung or gut damage and worsening of renal disease. Although hypoxia and HIF activation are present in several scenarios of renal diseases, further investigations are needed to clarify whether interfering with the HIF pathway is beneficial in different pathological contexts.
Collapse
Affiliation(s)
- Orestes Foresto-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Division of Nephrology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | | | - Marcella Cipelli
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Niels Olsen Saraiva Camara
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Division of Nephrology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Li K, Mei X, Xu K, Jia L, Zhao P, Tian Y, Li J. Comparative study of cigarette smoke, Klebsiella pneumoniae, and their combination on airway epithelial barrier function in mice. ENVIRONMENTAL TOXICOLOGY 2023; 38:1133-1142. [PMID: 36757011 DOI: 10.1002/tox.23753] [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: 10/04/2022] [Revised: 01/09/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The airway epithelium acts as a physical barrier to protect pulmonary airways against pathogenic microorganisms and toxic substances, such as cigarette smoke (CS), bacteria, and viruses. The disruption of the structural integrity and dysfunction of the airway epithelium is related to the occurrence and progression of chronic obstructive pulmonary disease. PURPOSE The aim of this study is to compare the effects of CS, Klebsiella pneumoniae (KP), and their combination on airway epithelial barrier function. METHODS The mice were exposed to CS, KP, and their combination from 1 to 8 weeks. After the cessation of CS and KP at Week 8, we observed the recovery of epithelial barrier function in mice for an additional 16 weeks. To compare the epithelial barrier function among different groups over time, the mice were sacrificed at Weeks 4, 8, 16, and 24 and then the lungs were harvested to detect the pulmonary pathology, inflammatory cytokines, and tight junction proteins. To determine the underlying mechanisms, the BEAS-2B cells were treated with an epidermal growth factor receptor (EGFR) inhibitor (AG1478). RESULTS The results of this study suggested that the decreased lung function, increased bronchial wall thickness (BWT), elevated inflammatory factors, and reduced tight junction protein levels were observed at Week 8 in CS-induced mice and these changes persisted until Week 16. In the KP group, increased BWT and elevated inflammatory factors were observed only at Week 8, whereas in the CS + KP group, decreased lung function, lung tissue injury, inflammatory cell infiltration, and epithelial barrier impairment were observed at Week 4 and persisted until Week 24. To further determine the mechanisms of CS, bacteria, and their combination on epithelial barrier injury, we investigated the changes of EGFR and its downstream protein in the lung tissues of mice and BEAS-2B cells. Our research indicated that CS, KP, or their combination could activate EGFR, which can phosphorylate and activate ERK1/2, and this effect was more pronounced in the CS + KP group. Furthermore, the EGFR inhibitor AG1478 suppressed the phosphorylation of ERK1/2 and subsequently upregulated the expression of ZO-1 and occludin. In general, these results indicated that the combination of CS and KP caused more severe and enduring damage to epithelial barrier function than CS or KP alone, which might be associated with EGFR/ERK1/2 signaling. CONCLUSION Epithelial barrier injury occurred earlier, was more severe, and had a longer duration when induced by the combination of CS and KP compared with the exposure to CS or KP alone, which might be associated with EGFR/ERK signaling.
Collapse
Affiliation(s)
- Kangchen Li
- Department of Respiratory Diseases, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaofeng Mei
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Kexin Xu
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lidan Jia
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Peng Zhao
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yange Tian
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jiansheng Li
- Department of Respiratory Diseases, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-constructed by Henan Province & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, China
- Department of Respiratory Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
4
|
Li X, Ma TK, Wang M, Zhang XD, Liu TY, Liu Y, Huang ZH, Zhu YH, Zhang S, Yin L, Xu YY, Ding H, Liu C, Shi H, Fan QL. YY1-induced upregulation of LncRNA-ARAP1-AS2 and ARAP1 promotes diabetic kidney fibrosis via aberrant glycolysis associated with EGFR/PKM2/HIF-1α pathway. Front Pharmacol 2023; 14:1069348. [PMID: 36874012 PMCID: PMC9974832 DOI: 10.3389/fphar.2023.1069348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Objectives: Dimeric pyruvate kinase (PK) M2 (PKM2) plays an important role in promoting the accumulation of hypoxia-inducible factor (HIF)-1α, mediating aberrant glycolysis and inducing fibrosis in diabetic kidney disease (DKD). The aim of this work was to dissect a novel regulatory mechanism of Yin and Yang 1 (YY1) on lncRNA-ARAP1-AS2/ARAP1 to regulate EGFR/PKM2/HIF-1α pathway and glycolysis in DKD. Materials and methods: We used adeno-associated virus (AAV)-ARAP1 shRNA to knocked down ARAP1 in diabetic mice and overexpressed or knocked down YY1, ARAP1-AS2 and ARAP1 expression in human glomerular mesangial cells. Gene levels were assessed by Western blotting, RT-qPCR, immunofluorescence staining and immunohistochemistry. Molecular interactions were determined by RNA pull-down, co-immunoprecipitation, ubiquitination assay and dual-luciferase reporter analysis. Results: YY1, ARAP1-AS2, ARAP1, HIF-1α, glycolysis and fibrosis genes expressions were upregulated and ARAP1 knockdown could inhibit dimeric PKM2 expression and partly restore tetrameric PKM2 formation, while downregulate HIF-1α accumulation and aberrant glycolysis and fibrosis in in-vivo and in-vitro DKD models. ARAP1 knockdown attenuates renal injury and renal dysfunction in diabetic mice. ARAP1 maintains EGFR overactivation in-vivo and in-vitro DKD models. Mechanistically, YY1 transcriptionally upregulates ARAP1-AS2 and indirectly regulates ARAP1 and subsequently promotes EGFR activation, HIF-1α accumulation and aberrant glycolysis and fibrosis. Conclusion: Our results first highlight the role of the novel regulatory mechanism of YY1 on ARAP1-AS2 and ARAP1 in promoting aberrant glycolysis and fibrosis by EGFR/PKM2/HIF-1α pathway in DKD and provide potential therapeutic strategies for DKD treatments.
Collapse
Affiliation(s)
- Xin Li
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Tian-Kui Ma
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Min Wang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Xiao-Dan Zhang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Tian-Yan Liu
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Yue Liu
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Zhao-Hui Huang
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Yong-Hong Zhu
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
| | - Shuang Zhang
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Li Yin
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Yan-Yan Xu
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Hong Ding
- Department of Nephrology, Fourth Hospital of China Medical University, Shenyang, China
| | - Cong Liu
- Department of General Surgery, First Hospital of Harbin Medical University, Harbin, China
| | - Hang Shi
- Department of Intensive Care Unit, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Qiu-Ling Fan
- Department of Nephrology, First Hospital of China Medical University, Shenyang, China
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Tajbakhsh A, Gheibihayat SM, Mortazavi D, Medhati P, Rostami B, Savardashtaki A, Momtazi-Borojeni AA. The Effect of Cigarette Smoke Exposure on Efferocytosis in Chronic Obstructive Pulmonary Disease; Molecular Mechanisms and Treatment Opportunities. COPD 2021; 18:723-736. [PMID: 34865568 DOI: 10.1080/15412555.2021.1978419] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cigarette smoking-related inflammation, cellular stresses, and tissue destruction play a key role in lung disease, such as chronic obstructive pulmonary disease (COPD). Notably, augmented apoptosis and impaired clearance of apoptotic cells, efferocytosis, contribute to the chronic inflammatory response and tissue destruction in patients with COPD. Of note, exposure to cigarette smoke can impair alveolar macrophages efferocytosis activity, which leads to secondary necrosis formation and tissue inflammation. A better understanding of the processes behind the effect of cigarette smoke on efferocytosis concerning lung disorders can help to design more efficient treatment approaches and also delay the development of lung disease, such as COPD. To this end, we aimed to seek mechanisms underlying the impairing effect of cigarette smoke on macrophages-mediated efferocytosis in COPD. Further, available therapeutic opportunities for restoring efferocytosis activity and ameliorating respiratory tract inflammation in smokers with COPD were also discussed.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Deniz Mortazavi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Pourya Medhati
- Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Rostami
- Health & Treatment Center of Rostam, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Abbas Momtazi-Borojeni
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Iran's National Elites Foundation, Tehran, Iran
| |
Collapse
|
6
|
Liu M, Wu K, Lin J, Xie Q, Liu Y, Huang Y, Zeng J, Yang Z, Wang Y, Dong S, Deng W, Yang M, Wu S, Jiang W, Li X. Emerging Biological Functions of IL-17A: A New Target in Chronic Obstructive Pulmonary Disease? Front Pharmacol 2021; 12:695957. [PMID: 34305606 PMCID: PMC8294190 DOI: 10.3389/fphar.2021.695957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/23/2021] [Indexed: 01/10/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease that causes high rates of disability and mortality worldwide because of severe progressive and irreversible symptoms. During the period of COPD initiation and progression, the immune system triggers the activation of various immune cells, including Regulatory T cells (Tregs), dendritic cells (DCs) and Th17 cells, and also the release of many different cytokines and chemokines, such as IL-17A and TGF-β. In recent years, studies have focused on the role of IL-17A in chronic inflammation process, which was found to play a highly critical role in facilitating COPD. Specially, IL-17A and its downstream regulators are potential therapeutic targets for COPD. We mainly focused on the possibility of IL-17A signaling pathways that involved in the progression of COPD; for instance, how IL-17A promotes airway remodeling in COPD? How IL-17A facilitates neutrophil inflammation in COPD? How IL-17A induces the expression of TSLP to promote the progression of COPD? Whether the mature DCs and Tregs participate in this process and how they cooperate with IL-17A to accelerate the development of COPD? And above associated studies could benefit clinical application of therapeutic targets of the disease. Moreover, four novel efficient therapies targeting IL-17A and other molecules for COPD are also concluded, such as Bufei Yishen formula (BYF), a Traditional Chinese Medicine (TCM), and curcumin, a natural polyphenol extracted from the root of Curcuma longa.
Collapse
Affiliation(s)
- Meiling Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Kang Wu
- Shenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China.,South China Hospital, Shenzhen University, Shenzhen, China
| | - Jinduan Lin
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Qingqiang Xie
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuan Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yin Huang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jun Zeng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhaogang Yang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yifan Wang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Shiyan Dong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Weiye Deng
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mingming Yang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Song Wu
- Shenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China.,South China Hospital, Shenzhen University, Shenzhen, China
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xuefeng Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital; State Key Laboratory of Respiratory Disease, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China.,Shenzhen Luohu People's Hospital, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
7
|
Carlier FM, de Fays C, Pilette C. Epithelial Barrier Dysfunction in Chronic Respiratory Diseases. Front Physiol 2021; 12:691227. [PMID: 34248677 PMCID: PMC8264588 DOI: 10.3389/fphys.2021.691227] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
Mucosal surfaces are lined by epithelial cells, which provide a complex and adaptive module that ensures first-line defense against external toxics, irritants, antigens, and pathogens. The underlying mechanisms of host protection encompass multiple physical, chemical, and immune pathways. In the lung, inhaled agents continually challenge the airway epithelial barrier, which is altered in chronic diseases such as chronic obstructive pulmonary disease, asthma, cystic fibrosis, or pulmonary fibrosis. In this review, we describe the epithelial barrier abnormalities that are observed in such disorders and summarize current knowledge on the mechanisms driving impaired barrier function, which could represent targets of future therapeutic approaches.
Collapse
Affiliation(s)
- François M. Carlier
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology and Lung Transplant, Centre Hospitalier Universitaire UCL Namur, Yvoir, Belgium
| | - Charlotte de Fays
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Charles Pilette
- Pole of Pneumology, ENT, and Dermatology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
- Department of Pneumology, Cliniques universitaires St-Luc, Brussels, Belgium
| |
Collapse
|
8
|
Moradi S, Jarrahi E, Ahmadi A, Salimian J, Karimi M, Zarei A, Azimzadeh Jamalkandi S, Ghanei M. PI3K signalling in chronic obstructive pulmonary disease and opportunities for therapy. J Pathol 2021; 254:505-518. [PMID: 33959951 DOI: 10.1002/path.5696] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/01/2021] [Accepted: 04/26/2021] [Indexed: 11/08/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease characterised by airway inflammation and progressive obstruction of the lung airflow. Current pharmacological treatments include bronchodilators, alone or in combination with steroids, or other anti-inflammatory agents, which have only partially contributed to the inhibition of disease progression and mortality. Therefore, further research unravelling the underlying mechanisms is necessary to develop new anti-COPD drugs with both lower toxicity and higher efficacy. Extrinsic signalling pathways play crucial roles in COPD development and exacerbations. In particular, phosphoinositide 3-kinase (PI3K) signalling has recently been shown to be a major driver of the COPD phenotype. Therefore, several small-molecule inhibitors have been identified to block the hyperactivation of this signalling pathway in COPD patients, many of them showing promising outcomes in both preclinical animal models of COPD and human clinical trials. In this review, we discuss the critically important roles played by hyperactivated PI3K signalling in the pathogenesis of COPD. We also critically review current therapeutics based on PI3K inhibition, and provide suggestions focusing on PI3K signalling for the further improvement of the COPD phenotype. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sharif Moradi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Esmaeil Jarrahi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ali Ahmadi
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Salimian
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Karimi
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Zarei
- Department of Traditional Medicine, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sadegh Azimzadeh Jamalkandi
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mostafa Ghanei
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Mucus composition abnormalities in sinonasal mucosa of chronic rhinosinusitis with and without nasal polyps. Inflammation 2021; 44:1937-1948. [PMID: 33999330 DOI: 10.1007/s10753-021-01471-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/11/2021] [Accepted: 04/15/2021] [Indexed: 12/30/2022]
Abstract
Mucus secretion and its composition are vital in the maintenance of airway health, among which hypoxia-inducible factors (HIFs) are thought to be involved in the regulation of mucin synthesis and regulation. Nasal mucus composition difference between healthy individuals and chronic rhinosinusitis (CRS) patients may contribute to the pathology of chronic nasal diseases, but so far, their role has yet to be completely understood. Nasal biopsy specimens were obtained from 24 healthy subjects and 99 patients with CRS without (CRSsNP, n=36) or with (CRSwNP, n=63) nasal polyps. Immunohistochemical (IHC) and immunofluorescent (IF) staining, quantitative real-time PCR, and western blot were performed to compare the nasal mucus composition between the subjects. Areas of the serous gland and mucous gland were both significantly increased in CRSsNP patients. In CRSwNP patients, a decrease in submucosal gland density and a marked increase in goblet cells were observed. The major gel-forming mucins in the sinonasal mucosa of CRSsNP and CRSwNP are MUC5B and MUC5AC respectively. Mucous cells are found in a higher proportion in both CRSsNP and CRSwNP. The proportion of MUC5AC-positive goblet cells was increased in CRSwNP. The mRNA level of HIF-2α was significantly increased in CRS, and both HIF-1α and HIF-2α were expressed in serous cell but not mucous cell. Over secretion and altered composition of mucus are observed in sinonasal mucosa of CRS, which was mainly associated with glandular hyperplasia in CRSsNP and goblet cell hyperplasia in CRSwNP. Mucus abnormality compromised both non-specific and specific antimicrobial capabilities in the sinonasal mucosa. HIF expression may contribute to differences in mucin synthesis and serous gland regulation, which needs further investigation to understand the pathology of CRS.
Collapse
|
10
|
Page LK, Staples KJ, Spalluto CM, Watson A, Wilkinson TMA. Influence of Hypoxia on the Epithelial-Pathogen Interactions in the Lung: Implications for Respiratory Disease. Front Immunol 2021; 12:653969. [PMID: 33868294 PMCID: PMC8044850 DOI: 10.3389/fimmu.2021.653969] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Under normal physiological conditions, the lung remains an oxygen rich environment. However, prominent regions of hypoxia are a common feature of infected and inflamed tissues and many chronic inflammatory respiratory diseases are associated with mucosal and systemic hypoxia. The airway epithelium represents a key interface with the external environment and is the first line of defense against potentially harmful agents including respiratory pathogens. The protective arsenal of the airway epithelium is provided in the form of physical barriers, and the production of an array of antimicrobial host defense molecules, proinflammatory cytokines and chemokines, in response to activation by receptors. Dysregulation of the airway epithelial innate immune response is associated with a compromised immunity and chronic inflammation of the lung. An increasing body of evidence indicates a distinct role for hypoxia in the dysfunction of the airway epithelium and in the responses of both innate immunity and of respiratory pathogens. Here we review the current evidence around the role of tissue hypoxia in modulating the host-pathogen interaction at the airway epithelium. Furthermore, we highlight the work needed to delineate the role of tissue hypoxia in the pathophysiology of chronic inflammatory lung diseases such as asthma, cystic fibrosis, and chronic obstructive pulmonary disease in addition to novel respiratory diseases such as COVID-19. Elucidating the molecular mechanisms underlying the epithelial-pathogen interactions in the setting of hypoxia will enable better understanding of persistent infections and complex disease processes in chronic inflammatory lung diseases and may aid the identification of novel therapeutic targets and strategies.
Collapse
Affiliation(s)
- Lee K. Page
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Karl J. Staples
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - C. Mirella Spalluto
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Alastair Watson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
- Birmingham Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Tom M. A. Wilkinson
- Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
- NIHR Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
11
|
Zhang Y, Wang Z, Zhang Y, Tong H, Zhang Y, Lu T. Potential Mechanisms for Traditional Chinese Medicine in Treating Airway Mucus Hypersecretion Associated With Coronavirus Disease 2019. Front Mol Biosci 2020; 7:577285. [PMID: 33381519 PMCID: PMC7768030 DOI: 10.3389/fmolb.2020.577285] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/27/2020] [Indexed: 01/08/2023] Open
Abstract
Background The rapid development of coronavirus disease 2019 (COVID-19) pandemic has become a great threat to global health. Its mortality is associated with inflammation-related airway mucus hypersecretion and dysfunction of expectoration, and the subsequent mucus blockage of the bronchioles at critical stage is attributed to hypoxemia, complications, and even death. Traditional Chinese medicine (TCM) has rich experience in expectorant, including treatment of COVID-19 patients with airway mucus dysfunction, yet little is known about the mechanisms. This study is aiming to explore the potential biological basis of TCM herbal expectorant for treating COVID-19. Objective To get core herbs with high used frequency applications in the actions of expectoration by using association rule algorithm and to investigate the multitarget mechanisms of core herbs in expectorant formulae for COVID-19 therapies. Methods Forty prescriptions for expectorant were retrieved from TCM Formulae. The ingredient compounds and targets of core herbs were collected from the TCMSP database, Gene-Cards, and NCBI. The protein interaction network (PPI) was constructed by SRING, and the network analysis was done by Cytoscape software. Bioconductor was applied for functional enrichment analysis of targets. Results The core herbs of expectorant could regulate core pathways (MAP kinase activity, cytokine receptor binding, G-protein-coupled receptor binding, etc.) via interactions of ingredients (glycyrol, citromitin, etc.) on mucin family to eliminate phlegm. Conclusion TCM herbal expectorant could regulate MAPK and cytokine-related pathways, thereby modulating Mucin-family to affect mucus generation and clearance and eventually retarding the deterioration of COVID-19 disease.
Collapse
Affiliation(s)
- Yuanfeng Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Zheyi Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China.,Department of Encephalopathy, Dongzhimen Hospital, Affiliated to BUCM, Beijing, China
| | - Yue Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hongxuan Tong
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yiling Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Tao Lu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
12
|
Szostak J, Titz B, Schlage WK, Guedj E, Sewer A, Phillips B, Leroy P, Buettner A, Neau L, Trivedi K, Martin F, Ivanov NV, Vanscheeuwijck P, Peitsch MC, Hoeng J. Structural, functional, and molecular impact on the cardiovascular system in ApoE -/- mice exposed to aerosol from candidate modified risk tobacco products, Carbon Heated Tobacco Product 1.2 and Tobacco Heating System 2.2, compared with cigarette smoke. Chem Biol Interact 2020; 315:108887. [PMID: 31705857 DOI: 10.1016/j.cbi.2019.108887] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/26/2022]
Abstract
AIM To investigate the molecular, structural, and functional impact of aerosols from candidate modified risk tobacco products (cMRTP), the Carbon Heated Tobacco Product (CHTP) 1.2 and Tobacco Heating System (THS) 2.2, compared with that of mainstream cigarette smoke (CS) on the cardiovascular system of ApoE-/- mice. METHODS Female ApoE-/- mice were exposed to aerosols from THS 2.2 and CHTP 1.2 or to CS from the 3R4F reference cigarette for up to 6 months at matching nicotine concentrations. A Cessation and a Switching group (3 months exposure to 3R4F CS followed by filtered air or CHTP 1.2 for 3 months) were included. Cardiovascular effects were investigated by echocardiographic, histopathological, immunohistochemical, and transcriptomics analyses. RESULTS Continuous exposure to cMRTP aerosols did not affect atherosclerosis progression, heart function, left ventricular (LV) structure, or the cardiovascular transcriptome. Exposure to 3R4F CS triggered atherosclerosis progression, reduced systolic ejection fraction and fractional shortening, caused heart LV hypertrophy, and initiated significant dysregulation in the transcriptomes of the heart ventricle and thoracic aorta. Importantly, the structural, functional, and molecular changes caused by 3R4F CS were improved in the smoking cessation and switching groups. CONCLUSION Exposure to cMRTP aerosols lacked most of the CS exposure-related functional, structural, and molecular effects. Smoking cessation or switching to CHTP 1.2 aerosol caused similar recovery from the 3R4F CS effects in the ApoE-/- model, with no further acceleration of plaque progression beyond the aging-related rate.
Collapse
Affiliation(s)
- Justyna Szostak
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Bjoern Titz
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Walter K Schlage
- Biology Consultant, Max-Baermann-Str. 21, 51429, Bergisch Gladbach, Germany.
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Alain Sewer
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Blaine Phillips
- PMI R&D, Philip Morris International Research Laboratories Pte. Ltd., Science Park II, Singapore.
| | - Patrice Leroy
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | | | - Laurent Neau
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Keyur Trivedi
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Florian Martin
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Nikolai V Ivanov
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Patrick Vanscheeuwijck
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Manuel C Peitsch
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A, Quai Jeanrenaud 5, CH-2000, Neuchâtel, Switzerland.
| |
Collapse
|
13
|
Zeglinski MR, Turner CT, Zeng R, Schwartz C, Santacruz S, Pawluk MA, Zhao H, Chan AWH, Carlsten C, Granville DJ. Soluble Wood Smoke Extract Promotes Barrier Dysfunction in Alveolar Epithelial Cells through a MAPK Signaling Pathway. Sci Rep 2019; 9:10027. [PMID: 31296909 PMCID: PMC6624307 DOI: 10.1038/s41598-019-46400-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 06/07/2019] [Indexed: 02/07/2023] Open
Abstract
Wildfire smoke induces acute pulmonary distress and is of particular concern to risk groups such as the sick and elderly. Wood smoke (WS) contains many of the same toxic compounds as those found in cigarette smoke (CS) including polycyclic aromatic hydrocarbons, carbon monoxide, and free radicals. CS is a well-established risk factor for respiratory diseases such as asthma and COPD. Limited studies investigating the biological effects of WS on the airway epithelium have been performed. Using a cell culture-based model, we assessed the effects of a WS-infused solution on alveolar epithelial barrier function, cell migration, and survival. The average geometric mean of particles in the WS was 178 nm. GC/MS analysis of the WS solution identified phenolic and cellulosic compounds. WS exposure resulted in a significant reduction in barrier function, which peaked after 24 hours of continuous exposure. The junctional protein E-cadherin showed a prominent reduction in response to increasing concentrations of WS. Furthermore, WS significantly repressed cell migration following injury to the cell monolayer. There was no difference in cell viability following WS exposure. Mechanistically, WS exposure induced activation of the p44/42, but not p38, MAPK signaling pathway, and inhibition of p44/42 phosphorylation prevented the disruption of barrier function and loss of E-cadherin staining. Thus, WS may contribute to the breakdown of alveolar structure and function through a p44/42 MAPK-dependent pathway and may lead to the development and/or exacerbation of respiratory pathologies with chronic exposure.
Collapse
Affiliation(s)
- Matthew R Zeglinski
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, University of British Columbia (UBC), Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, UBC, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, UBC, Vancouver, BC, Canada.,British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, BC, Canada
| | - Christopher T Turner
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, University of British Columbia (UBC), Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, UBC, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, UBC, Vancouver, BC, Canada.,British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, BC, Canada
| | - Rui Zeng
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Carley Schwartz
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, UBC, Vancouver, BC, Canada.,Department of Medicine, Division of Respiratory Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease, Vancouver Coastal Health Research Institute, UBC, Vancouver, BC, Canada
| | - Stephanie Santacruz
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, University of British Columbia (UBC), Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, UBC, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, UBC, Vancouver, BC, Canada.,British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, BC, Canada
| | - Megan A Pawluk
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Hongyan Zhao
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, University of British Columbia (UBC), Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, UBC, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, UBC, Vancouver, BC, Canada.,British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, BC, Canada
| | - Arthur W H Chan
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Christopher Carlsten
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, UBC, Vancouver, BC, Canada.,Department of Medicine, Division of Respiratory Medicine, Chan-Yeung Centre for Occupational and Environmental Respiratory Disease, Vancouver Coastal Health Research Institute, UBC, Vancouver, BC, Canada
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute, University of British Columbia (UBC), Vancouver, BC, Canada. .,Department of Pathology and Laboratory Medicine, UBC, Vancouver, BC, Canada. .,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, UBC, Vancouver, BC, Canada. .,British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, BC, Canada.
| |
Collapse
|
14
|
Lee JW, Ko J, Ju C, Eltzschig HK. Hypoxia signaling in human diseases and therapeutic targets. Exp Mol Med 2019; 51:1-13. [PMID: 31221962 PMCID: PMC6586801 DOI: 10.1038/s12276-019-0235-1] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023] Open
Abstract
Since the discovery of hypoxia-inducible factor (HIF), numerous studies on the hypoxia signaling pathway have been performed. The role of HIF stabilization during hypoxia has been extended from the induction of a single gene erythropoietin to the upregulation of a couple of hundred downstream targets, which demonstrates the complexity and importance of the HIF signaling pathway. Accordingly, HIF and its downstream targets are emerging as novel therapeutic options to treat various organ injuries. In this review, we discuss the current understanding of HIF signaling in four different organ systems, including the heart, lung, liver, and kidney. We also discuss the divergent roles of HIF in acute and chronic disease conditions and their revealed functions. Finally, we introduce some of the efforts that are being performed to translate our current knowledge in hypoxia signaling to clinical medicine.
Collapse
Affiliation(s)
- Jae W Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Junsuk Ko
- Department of Biochemistry and Molecular Biology, MD Anderson UT Health Graduate School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Cynthia Ju
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA
| | - Holger K Eltzschig
- Department of Anesthesiology, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
15
|
Liang HX, Sun JJ, Shen ZB, Yu BW, Cui HH, Yin YQ. A novel alkaloid glycoside isolated from Atalantia buxifolia. Nat Prod Res 2019; 34:3042-3047. [PMID: 31140296 DOI: 10.1080/14786419.2019.1607854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
From the dried root and rhizomes of Atalantia buxifolia afford 11 compounds, one new compound 1 and ten known compounds 2-11 were isolated and identified. The novel compound 1 was alkaloid glycoside. Its mother nuclear was the acridone, which was relatively rare. The structure of compound 1 was established identified by spectrum and elucidated as β-D-Glu-4,5-dimethoxy-1,6-dihydroxy-10-methyl-acridone. The compound 1 enriched the compound library and laid the material foundation for the subsequent study of pharmacological activities.
Collapse
Affiliation(s)
- Hui-Xian Liang
- School of Traditional Chinese Medicinal Chemistry, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jing-Jing Sun
- School of Traditional Chinese Medicinal Chemistry, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhi-Bin Shen
- School of Traditional Chinese Medicinal Chemistry, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bang-Wei Yu
- School of Traditional Chinese Medicinal Chemistry, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hong-Hua Cui
- School of Traditional Chinese Medicinal Chemistry, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yong-Qin Yin
- School of Traditional Chinese Medicinal Chemistry, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
16
|
Krishn SR, Ganguly K, Kaur S, Batra SK. Ramifications of secreted mucin MUC5AC in malignant journey: a holistic view. Carcinogenesis 2019; 39:633-651. [PMID: 29415129 DOI: 10.1093/carcin/bgy019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 02/01/2018] [Indexed: 12/14/2022] Open
Abstract
Heavily glycosylated secreted mucin MUC5AC, by the virtue of its cysteine-rich repeats, can form inter- and intramolecular disulfide linkages resulting in complex polymers, which in turn craft the framework of the polymeric mucus gel on epithelial cell surfaces. MUC5AC is a molecule with versatile functional implications including barrier functions to epithelial cells, host-pathogen interaction, immune cell attraction to sites of premalignant or malignant lesions and tumor progression in a context-dependent manner. Differential expression, glycosylation and localization of MUC5AC have been associated with a plethora of benign and malignant pathologies. In this era of robust technologies, overexpression strategies and genetically engineered mouse models, MUC5AC is emerging as a potential diagnostic, prognostic and therapeutic target for various malignancies. Considering the clinical relevance of MUC5AC, this review holistically encompasses its genomic organization, domain structure, glycosylation patterns, regulation, functional and molecular connotation from benign to malignant pathologies. Furthermore, we have here explored the incipient and significant experimental tools that are being developed to study this structurally complex and evolutionary conserved gel-forming mucin.
Collapse
Affiliation(s)
- Shiv Ram Krishn
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
17
|
Guan R, Wang J, Li Z, Ding M, Li D, Xu G, Wang T, Chen Y, Yang Q, Long Z, Cai Z, Zhang C, Liang X, Dong L, Zhao L, Zhang H, Sun D, Lu W. Sodium Tanshinone IIA Sulfonate Decreases Cigarette Smoke-Induced Inflammation and Oxidative Stress via Blocking the Activation of MAPK/HIF-1α Signaling Pathway. Front Pharmacol 2018; 9:263. [PMID: 29765317 PMCID: PMC5938387 DOI: 10.3389/fphar.2018.00263] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 03/09/2018] [Indexed: 12/27/2022] Open
Abstract
Aberrant activation of hypoxia-inducible factor (HIF)-1α is frequently encountered and promotes oxidative stress and inflammation in chronic obstructive pulmonary disease (COPD). The present study investigated whether sodium tanshinone IIA sulfonate (STS), a water-soluble derivative of tanshinone IIA, can mediate its effect through inhibiting HIF-1α–induced oxidative stress and inflammation in cigarette smoke (CS)-induced COPD in mice. Here, we found that STS improved pulmonary function, ameliorated emphysema and decreased the infiltration of inflammatory cells in the lungs of CS-exposed mice. STS reduced CS- and cigarette smoke extract (CSE)-induced upregulation of tumor necrosis factor (TNF)-α and interleukin (IL)-1β in the lungs and macrophages. STS also inhibited CSE-induced reactive oxygen species (ROS) production, as well as the upregulation of heme oxygenase (HO)-1, NOX1 and matrix metalloproteinase (MMP)-9 in macrophages. In addition, STS suppressed HIF-1α expression in vivo and in vitro, and pretreatment with HIF-1α siRNA reduced CSE-induced elevation of TNF-α, IL-1β, and HO-1 content in the macrophages. Moreover, we found that STS inhibited CSE-induced the phosphorylation of ERK, p38 MAPK and JNK in macrophages, and inhibition of these signaling molecules significantly repressed CSE-induced HIF-1α expression. It indicated that STS inhibits CSE-induced HIF-1α expression likely by blocking MAPK signaling. Furthermore, STS also promoted HIF-1α protein degradation in CSE-stimulated macrophages. Taken together, these results suggest that STS prevents COPD development possibly through the inhibition of HIF-1α signaling, and may be a novel strategy for the treatment of COPD.
Collapse
Affiliation(s)
- Ruijuan Guan
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jian Wang
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ziying Li
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Mingjing Ding
- Departments of Respiratory and Critical Diseases, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Defu Li
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guihua Xu
- Department of Clinical Medical Research Center, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Tao Wang
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuqin Chen
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qian Yang
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhen Long
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhou Cai
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chenting Zhang
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xue Liang
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Lian Dong
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Li Zhao
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haiyun Zhang
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Dejun Sun
- Departments of Respiratory and Critical Diseases, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Wenju Lu
- State Key Lab of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
18
|
Savin Z, Kivity S, Yonath H, Yehuda S. Smoking and the intestinal microbiome. Arch Microbiol 2018; 200:677-684. [DOI: 10.1007/s00203-018-1506-2] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 03/07/2018] [Accepted: 03/23/2018] [Indexed: 12/21/2022]
|
19
|
Lakshmi SP, Reddy AT, Reddy RC. Emerging pharmaceutical therapies for COPD. Int J Chron Obstruct Pulmon Dis 2017; 12:2141-2156. [PMID: 28790817 PMCID: PMC5531723 DOI: 10.2147/copd.s121416] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
COPD, for which cigarette smoking is the major risk factor, remains a worldwide burden. Current therapies provide only limited short-term benefit and fail to halt progression. A variety of potential therapeutic targets are currently being investigated, including COPD-related proinflammatory mediators and signaling pathways. Other investigational compounds target specific aspects or complications of COPD such as mucus hypersecretion and pulmonary hypertension. Although many candidate therapies have shown no significant effects, other emerging therapies have improved lung function, pulmonary hypertension, glucocorticoid sensitivity, and/or the frequency of exacerbations. Among these are compounds that inhibit the CXCR2 receptor, mitogen-activated protein kinase/Src kinase, myristoylated alanine-rich C kinase substrate, selectins, and the endothelin receptor. Activation of certain transcription factors may also be relevant, as a large retrospective cohort study of COPD patients with diabetes found that the peroxisome proliferator-activated receptor γ (PPARγ) agonists rosiglitazone and pioglitazone were associated with reduced COPD exacerbation rate. Notably, several therapies have shown efficacy only in identifiable subgroups of COPD patients, suggesting that subgroup identification may become more important in future treatment strategies. This review summarizes the status of emerging therapeutic pharmaceuticals for COPD and highlights those that appear most promising.
Collapse
Affiliation(s)
- Sowmya P Lakshmi
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Aravind T Reddy
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Raju C Reddy
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh School of Medicine.,Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Patel N, Trumph CD, Bodas M, Vij N. Role of second-hand smoke (SHS)-induced proteostasis/autophagy impairment in pediatric lung diseases. Mol Cell Pediatr 2017; 4:3. [PMID: 28150141 PMCID: PMC5289127 DOI: 10.1186/s40348-017-0069-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/02/2017] [Indexed: 12/11/2022] Open
Abstract
Background Exposure to second-hand tobacco smoke (SHS) is one of the prime risk factors for chronic lung disease development. Smoking during pregnancy may lead to birth defects in the newborn that include pulmonary dysfunction, increased susceptibility to opportunistic pathogens, or initiation of childhood respiratory manifestations such as bronchopulmonary dysplasia (BPD). Moreover, exposure to SHS in early childhood can have negative impact on lung health, although the exact mechanisms are unclear. Autophagy is a crucial proteostatic mechanism modulated by cigarette smoke (CS) in adult lungs. Here, we sought to investigate whether SHS exposure impairs autophagy in pediatric lungs. Methods Pregnant C57BL/6 mice were exposed to room air or SHS for 14 days. The newborn pups were subsequently exposed to room air or SHS (5 h/day) for 1 or 14 days, and lungs were harvested. Soluble and insoluble protein fractions isolated from pediatric mice lungs were subjected to immunoblotting for ubiquitin (Ub), p62, VCP, HIF-1α, and β-actin. Results Our data shows that short-term exposure to SHS (1 or 14 days) leads to proteostasis and autophagy-impairment as evident by significant increase in accumulation of ubiquitinated proteins (Ub), p62 (impaired-autophagy marker) and valosin-containing protein (VCP) in the insoluble protein fractions of pediatric mice lungs. Moreover, increased HIF-1α levels in SHS-exposed mice lungs points towards a novel mechanism for SHS-induced lung disease initiation in the pediatric population. Validating the in vivo studies, we demonstrate that treatment of human bronchial epithelial cells (Beas2b cells) with the proteasome inhibitor (MG-132) induces HIF-1α expression that is controlled by co-treatment with autophagy-inducing drug, cysteamine. Conclusions SHS-exposure induced proteostasis/autophagy impairment can mediate the initiation of chronic lung disease in pediatric subjects. Hence, our data warrants the evaluation of proteostasis/autophagy-inducing drugs, such as cysteamine, as a potential therapeutic intervention strategy for SHS-induced pediatric lung diseases.
Collapse
Affiliation(s)
- Neel Patel
- College of Medicine, Central Michigan University, College of Medicine Research Building, 2630 Denison Drive, Room# 120, Room# 120 (Office) and 126-127 (Lab), Mt Pleasant, MI, 48859, USA
| | - Christopher D Trumph
- College of Medicine, Central Michigan University, College of Medicine Research Building, 2630 Denison Drive, Room# 120, Room# 120 (Office) and 126-127 (Lab), Mt Pleasant, MI, 48859, USA
| | - Manish Bodas
- College of Medicine, Central Michigan University, College of Medicine Research Building, 2630 Denison Drive, Room# 120, Room# 120 (Office) and 126-127 (Lab), Mt Pleasant, MI, 48859, USA
| | - Neeraj Vij
- College of Medicine, Central Michigan University, College of Medicine Research Building, 2630 Denison Drive, Room# 120, Room# 120 (Office) and 126-127 (Lab), Mt Pleasant, MI, 48859, USA. .,Department of Pediatric Respiratory Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Roche O, Deguiz ML, Tiana M, Galiana-Ribote C, Martinez-Alcazar D, Rey-Serra C, Ranz-Ribeiro B, Casitas R, Galera R, Fernández-Navarro I, Sánchez-Cuéllar S, Bernard V, Ancochea J, Wasserman WW, García-Rio F, Jimenez B, Del Peso L. Identification of non-coding genetic variants in samples from hypoxemic respiratory disease patients that affect the transcriptional response to hypoxia. Nucleic Acids Res 2016; 44:9315-9330. [PMID: 27625398 PMCID: PMC5100585 DOI: 10.1093/nar/gkw811] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/02/2016] [Indexed: 12/11/2022] Open
Abstract
A wide range of diseases course with an unbalance between the consumption of oxygen by tissues and its supply. This situation triggers a transcriptional response, mediated by the hypoxia inducible factors (HIFs), that aims to restore oxygen homeostasis. Little is known about the inter-individual variation in this response and its role in the progression of disease. Herein, we sought to identify common genetic variants mapping to hypoxia response elements (HREs) and characterize their effect on transcription. To this end, we constructed a list of genome-wide HIF-binding regions from publicly available experimental datasets and studied the genetic variability in these regions by targeted re-sequencing of genomic samples from 96 chronic obstructive pulmonary disease and 144 obstructive sleep apnea patients. This study identified 14 frequent variants disrupting potential HREs. The analysis of the genomic regions containing these variants by means of reporter assays revealed that variants rs1009329, rs6593210 and rs150921338 impaired the transcriptional response to hypoxia. Finally, using genome editing we confirmed the functional role of rs6593210 in the transcriptional regulation of EGFR. In summary, we found that inter-individual variability in non-coding regions affect the response to hypoxia and could potentially impact on the progression of pulmonary diseases.
Collapse
Affiliation(s)
- Olga Roche
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain.,IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain.,Unidad de Medicina Molecular, laboratorio de Oncología, CRIB. Universidad de Castilla-La Mancha, 02006, Albacete, Spain
| | - María Laura Deguiz
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain.,IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain
| | - María Tiana
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain.,IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Clara Galiana-Ribote
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| | - Daniel Martinez-Alcazar
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| | - Carlos Rey-Serra
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| | - Beatriz Ranz-Ribeiro
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain
| | - Raquel Casitas
- IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain.,Hospital Universitario La Paz, IdiPAZ, Servicio de Neumología, 28029 Madrid, Spain
| | - Raúl Galera
- IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain.,Hospital Universitario La Paz, IdiPAZ, Servicio de Neumología, 28029 Madrid, Spain
| | - Isabel Fernández-Navarro
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain.,Hospital Universitario La Paz, IdiPAZ, Servicio de Neumología, 28029 Madrid, Spain
| | - Silvia Sánchez-Cuéllar
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain.,Servicio de Neumología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria del hospital de La Princesa, 28006 Madrid, Spain
| | - Virginie Bernard
- Departamento de Medicina, Universidad Autónoma de Madrid (UAM) 28029 Madrid, Spain
| | - Julio Ancochea
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain.,Servicio de Neumología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria del hospital de La Princesa, 28006 Madrid, Spain.,Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia Vancouver, British Columbia V5Z 4H4, Canada
| | - Wyeth W Wasserman
- Departamento de Medicina, Universidad Autónoma de Madrid (UAM) 28029 Madrid, Spain
| | - Francisco García-Rio
- IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain.,Hospital Universitario La Paz, IdiPAZ, Servicio de Neumología, 28029 Madrid, Spain.,Centre for Molecular Medicine and Therapeutics, Child and Family Research Institute, Department of Medical Genetics, University of British Columbia Vancouver, British Columbia V5Z 4H4, Canada
| | - Benilde Jimenez
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain.,IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luis Del Peso
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas 'Alberto Sols' (CSIC-UAM), 28029 Madrid, Spain .,IdiPaz, Instituto de Investigación Sanitaria del Hospital Universitario La Paz, 28029 Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
22
|
Ha EVS, Rogers DF. Novel Therapies to Inhibit Mucus Synthesis and Secretion in Airway Hypersecretory Diseases. Pharmacology 2015; 97:84-100. [PMID: 26674354 DOI: 10.1159/000442794] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/26/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND In asthma and chronic obstructive pulmonary disease (COPD), airway mucus hypersecretion contributes to impaired mucociliary clearance, mucostasis and, potentially, the development of mucus plugging of the airways. SUMMARY Excess mucus production can be targeted via therapies that focus on inhibition mucin synthesis, via reducing expression of mucin (MUC) genes, and/or inhibition of mucin secretion into the airways. KEY MESSAGES This review discusses a number of therapeutic approaches to reduce airway mucus in asthma and COPD, including the use of synthetic and natural products. In particular, it highlights areas where clinical trials of inhibitors of particular target molecules are lacking. Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors are an example of a targeted therapy that has been researched to reduce mucus synthesis, as have inhibitors of EGFR's downstream signalling pathways, for example, mitogen-activated protein kinase-13 and hypoxia inducible factor-1. However, their efficacy and safety profiles are currently not up to the mark. There is clinical potential in Bio-11006, which reduces mucus secretion via the inhibition of myristoylated alanine-rich C-kinase substrate and is currently in Phase IIb trial.
Collapse
Affiliation(s)
- Emily V S Ha
- National Heart and Lung Institute, Imperial College, London, UK
| | | |
Collapse
|
23
|
Allais L, Kerckhof FM, Verschuere S, Bracke KR, De Smet R, Laukens D, Van den Abbeele P, De Vos M, Boon N, Brusselle GG, Cuvelier CA, Van de Wiele T. Chronic cigarette smoke exposure induces microbial and inflammatory shifts and mucin changes in the murine gut. Environ Microbiol 2015; 18:1352-63. [PMID: 26033517 DOI: 10.1111/1462-2920.12934] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 05/28/2015] [Indexed: 12/21/2022]
Abstract
Inflammatory bowel diseases (IBD) are complex multifactorial diseases characterized by an inappropriate host response to an altered commensal microbiome and dysfunctional mucus barrier. Cigarette smoking is the best known environmental risk factor in IBD. Here, we studied the influence of chronic smoke exposure on the gut microbiome, mucus layer composition and immune factors in conventional mice. We compared smoke-exposed with air-exposed mice (n = 12) after a smoke exposure of 24 weeks. Both Illumina sequencing (n = 6) and denaturing gradient gel electrophoresis (n = 12) showed that bacterial activity and community structure were significantly altered in the colon due to smoke exposure. Interestingly, an increase of Lachnospiraceae sp. activity in the colon was observed. Also, the mRNA expression of Muc2 and Muc3 increased in the ileum, whereas Muc4 increased in the distal colon of smoke-exposed mice (n = 6). Furthermore, we observed increased Cxcl2 and decreased Ifn-γ in the ileum, and increased Il-6 and decreased Tgf-β in the proximal colon. Tight junction gene expression remained unchanged. We infer that the modulating role of chronic smoke exposure as a latently present risk factor in the gut may be driven by the altered epithelial mucus profiles and changes in microbiome composition and immune factors.
Collapse
Affiliation(s)
- Liesbeth Allais
- Department of Medical and Forensic Pathology, Ghent University, Ghent, Belgium
| | - Frederiek-Maarten Kerckhof
- Laboratory of Microbial Ecology and Technology (LabMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | | | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Rebecca De Smet
- Department of Medical and Forensic Pathology, Ghent University, Ghent, Belgium
| | - Debby Laukens
- Department of Gastroenterology, Ghent University, Ghent, Belgium
| | - Pieter Van den Abbeele
- Laboratory of Microbial Ecology and Technology (LabMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Martine De Vos
- Department of Gastroenterology, Ghent University, Ghent, Belgium
| | - Nico Boon
- Laboratory of Microbial Ecology and Technology (LabMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Guy G Brusselle
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Claude A Cuvelier
- Department of Medical and Forensic Pathology, Ghent University, Ghent, Belgium
| | - Tom Van de Wiele
- Laboratory of Microbial Ecology and Technology (LabMET), Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
24
|
Li R, Uttarwar L, Gao B, Charbonneau M, Shi Y, Chan JSD, Dubois CM, Krepinsky JC. High Glucose Up-regulates ADAM17 through HIF-1α in Mesangial Cells. J Biol Chem 2015; 290:21603-14. [PMID: 26175156 DOI: 10.1074/jbc.m115.651604] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Indexed: 12/26/2022] Open
Abstract
We previously showed that ADAM17 mediates high glucose-induced matrix production by kidney mesangial cells. ADAM17 expression is increased in diabetic kidneys, suggesting that its up-regulation may augment high glucose profibrotic responses. We thus studied the effects of high glucose on ADAM17 gene regulation. Primary rat mesangial cells were treated with high glucose (30 mm) or mannitol as osmotic control. High glucose dose-dependently increased ADAM17 promoter activity, transcript, and protein levels. This correlated with augmented ADAM17 activity after 24 h versus 1 h of high glucose. We tested involvement of transcription factors shown in other settings to regulate ADAM17 transcription. Promoter activation was not affected by NF-κB or Sp1 inhibitors, but was blocked by hypoxia-inducible factor-1α (HIF-1α) inhibition or down-regulation. This also prevented ADAM17 transcript and protein increases. HIF-1α activation by high glucose was shown by its increased nuclear translocation and activation of the HIF-responsive hypoxia-response element (HRE)-luciferase reporter construct. Assessment of ADAM17 promoter deletion constructs coupled with mutation analysis and ChIP studies identified HIF-1α binding to its consensus element at -607 as critical for the high glucose response. Finally, inhibitors of epidermal growth factor receptor (EGFR) and downstream PI3K/Akt, or ADAM17 itself, prevented high glucose-induced HIF-1α activation and ADAM17 up-regulation. Thus, high glucose induces ADAM17 transcriptional up-regulation in mesangial cells, which is associated with augmentation of its activity. This is mediated by HIF-1α and requires EGFR/ADAM17 signaling, demonstrating the potentiation by ADAM17 of its own up-regulation. ADAM17 inhibition thus provides a potential novel therapeutic strategy for the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Renzhong Li
- From the Division of Nephrology, McMaster University, Hamilton, Ontario L8N 4A6
| | - Lalita Uttarwar
- From the Division of Nephrology, McMaster University, Hamilton, Ontario L8N 4A6
| | - Bo Gao
- From the Division of Nephrology, McMaster University, Hamilton, Ontario L8N 4A6
| | - Martine Charbonneau
- the Division of Immunology, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, and
| | - Yixuan Shi
- the Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - John S D Chan
- the Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Quebec H2X 0A9, Canada
| | - Claire M Dubois
- the Division of Immunology, Université de Sherbrooke, Sherbrooke, Quebec J1H 5N4, and
| | - Joan C Krepinsky
- From the Division of Nephrology, McMaster University, Hamilton, Ontario L8N 4A6,
| |
Collapse
|
25
|
Kanai K, Koarai A, Shishikura Y, Sugiura H, Ichikawa T, Kikuchi T, Akamatsu K, Hirano T, Nakanishi M, Matsunaga K, Minakata Y, Ichinose M. Cigarette smoke augments MUC5AC production via the TLR3-EGFR pathway in airway epithelial cells. Respir Investig 2015; 53:137-48. [PMID: 26100173 DOI: 10.1016/j.resinv.2015.01.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 01/05/2015] [Accepted: 01/21/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND Viral infections are a major cause of chronic obstructive pulmonary disease (COPD) exacerbations. Toll-like receptor 3 (TLR3) reacts with double-stranded RNA (dsRNA) and participates in the immune response after viral infection. In the present study, we examined whether cigarette smoke, which is involved in the pathogenesis of COPD, enhances mucin production via the TLR3-epidermal growth factor receptor (EGFR) pathway in airway epithelial cells. METHODS We studied the effects of cigarette smoke extract (CSE) on signal transduction and the production of mucin 5AC (MUC5AC) in NCI-H292 cells and differentiated primary human bronchial epithelial cells stimulated with a synthetic dsRNA analogue, polyinosinic-polycytidylic acid [poly(I:C)], used as a TLR3 ligand. RESULTS CSE significantly potentiated the production of MUC5AC in epithelial cells stimulated with poly(I:C). Antibodies to EGFR or EGFR ligands inhibited CSE-augmented MUC5AC release in poly(I:C)-treated cells. Treatment with poly(I:C) or CSE alone increased the phosphorylation of EGFR and extracellular signal-regulated kinase (ERK). However, after poly(I:C) stimulation, CSE did not enhance EGFR phosphorylation, but did augment ERK phosphorylation. EGFR inhibitors and an ERK inhibitor inhibited the augmented release of MUC5AC. In addition, treatment with N-acetylcysteine, an antioxidant, inhibited the CSE-augmented phosphorylation of ERK and MUC5AC. CONCLUSIONS These data show that cigarette smoke increases TLR3-stimulated MUC5AC production in airway epithelial cells, mainly via ERK signaling. The effect might be mediated in part by oxidative stress. Modulation of this pathway might be a therapeutic target for viral-induced mucin overproduction in COPD exacerbation.
Collapse
Affiliation(s)
- Kuninobu Kanai
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Akira Koarai
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Yutaka Shishikura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | - Tomohiro Ichikawa
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Takashi Kikuchi
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Keiichiro Akamatsu
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Tsunahiko Hirano
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Masanori Nakanishi
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Kazuto Matsunaga
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Yoshiaki Minakata
- Third Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan.
| | - Masakazu Ichinose
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| |
Collapse
|
26
|
Yu Q, Chen X, Fang X, Chen Q, Hu C. Caveolin-1 aggravates cigarette smoke extract-induced MUC5AC secretion in human airway epithelial cells. Int J Mol Med 2015; 35:1435-42. [PMID: 25776934 DOI: 10.3892/ijmm.2015.2133] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/04/2015] [Indexed: 11/06/2022] Open
Abstract
Airway mucus hypersecretion is a major pathological characteristic of chronic obstructive pulmonary disease (COPD), and cigarette smoke is highly implicated in mucus secretion and the development of COPD. Cigarette smoke reportedly induces mucin overproduction through the epidermal growth factor receptor (EGFR) in the airway epithelium; however, the underlying mechanisms responsible for the activation of EGFR remain unknown. Caveolin-1, a component protein in the cytomembrane, reportedly regulates airway inflammation and lung injury. In this study, we aimed to determine whether caveolin-1 modulates mucin hyperproduction induced by cigarette smoke. Our results revealed that cigarette smoke extract (CSE) significantly increased MUC5AC production, as well as the levels of phosphorylated EGFR (p-EGFR) and phosphorylated Akt (p-Akt) in human bronchial epithelial cells (16HBE cells), as shown by ELISA, RT-PCR and western blot analysis. These effects were prevented by treatment with EGFR inhibitor (AG1478) and phosphatidylinostol-3-kinase (PI3K) inhibitor (LY294002). We also found that the overexpression of caveolin-1 enhanced the expression of MUC5AC, p-EGFR and p-Akt induced by CSE. Conversely, the downregulation of caveolin-1 by siRNA against caveolin-1 inhibited the expression of MUC5AC, p-EGFR and p-Akt. Taken together, our data suggest that caveolin-1 enhances CSE-induced MUC5AC hypersecretion through the EGFR/PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Qiao Yu
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xi Chen
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xia Fang
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Qiong Chen
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Chengping Hu
- Department of Gerontology and Respirology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
27
|
Higashi T, Mai Y, Noya Y, Horinouchi T, Terada K, Hoshi A, Nepal P, Harada T, Horiguchi M, Hatate C, Kuge Y, Miwa S. A simple and rapid method for standard preparation of gas phase extract of cigarette smoke. PLoS One 2014; 9:e107856. [PMID: 25229830 PMCID: PMC4168273 DOI: 10.1371/journal.pone.0107856] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 08/15/2014] [Indexed: 12/19/2022] Open
Abstract
Cigarette smoke consists of tar and gas phase: the latter is toxicologically important because it can pass through lung alveolar epithelium to enter the circulation. Here we attempt to establish a standard method for preparation of gas phase extract of cigarette smoke (CSE). CSE was prepared by continuously sucking cigarette smoke through a Cambridge filter to remove tar, followed by bubbling it into phosphate-buffered saline (PBS). An increase in dry weight of the filter was defined as tar weight. Characteristically, concentrations of CSEs were represented as virtual tar concentrations, assuming that tar on the filter was dissolved in PBS. CSEs prepared from smaller numbers of cigarettes (original tar concentrations ≤ 15 mg/ml) showed similar concentration-response curves for cytotoxicity versus virtual tar concentrations, but with CSEs from larger numbers (tar ≥ 20 mg/ml), the curves were shifted rightward. Accordingly, the cytotoxic activity was detected in PBS of the second reservoir downstream of the first one with larger numbers of cigarettes. CSEs prepared from various cigarette brands showed comparable concentration-response curves for cytotoxicity. Two types of CSEs prepared by continuous and puff smoking protocols were similar regarding concentration-response curves for cytotoxicity, pharmacology of their cytotoxicity, and concentrations of cytotoxic compounds. These data show that concentrations of CSEs expressed by virtual tar concentrations can be a reference value to normalize their cytotoxicity, irrespective of numbers of combusted cigarettes, cigarette brands and smoking protocols, if original tar concentrations are ≤15 mg/ml.
Collapse
Affiliation(s)
- Tsunehito Higashi
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yosuke Mai
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yoichi Noya
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takahiro Horinouchi
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Koji Terada
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Akimasa Hoshi
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Prabha Nepal
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takuya Harada
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mika Horiguchi
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Chizuru Hatate
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Soichi Miwa
- Department of Cellular Pharmacology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- * E-mail:
| |
Collapse
|
28
|
Martin C, Frija-Masson J, Burgel PR. Targeting Mucus Hypersecretion: New Therapeutic Opportunities for COPD? Drugs 2014; 74:1073-89. [DOI: 10.1007/s40265-014-0235-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
29
|
Iwanaga K, Elliott MS, Vedal S, Debley JS. Urban particulate matter induces pro-remodeling factors by airway epithelial cells from healthy and asthmatic children. Inhal Toxicol 2014; 25:653-60. [PMID: 24102466 DOI: 10.3109/08958378.2013.827283] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONTEXT Chronic exposure to ambient particulate matter pollution during childhood is associated with decreased lung function growth and increased prevalence of reported respiratory symptoms. The role of airway epithelium-derived factors has not been well determined. OBJECTIVE To determine if urban particulate matter (UPM) stimulates production of vascular endothelial growth factor (VEGF) and transforming growth factor-β2 (TGF-β2), and gene expression of mucin 5AC (MUC5AC) and interleukin-(IL)-8 by primary airway epithelial cells (AECs) obtained from carefully phenotyped healthy and atopic asthmatic school-aged children. METHODS Primary AECs from 9 healthy and 14 asthmatic children were differentiated in air--liquid interface (ALI) culture. The apical surface was exposed to UPM suspension or phosphate buffered saline (PBS) vehicle control for 96 h. VEGF and TGF-β2 concentrations in cell media at baseline, 48 and 96 h were measured via ELISA. MUC5AC and IL-8 expression by AECs at 96 h was measured via quantitative polymerase chain reaction. RESULTS Baseline concentrations of VEGF, but not TGF-β2, were significantly higher in asthmatic versus healthy cultures. UPM stimulated production of VEGF, but not TGF-β2, at 48 and 96 h; the magnitude of change was comparable across groups. At 96 h there was greater MUC5AC and IL-8 expression by UPM exposed compared to PBS exposed AECs. CONCLUSIONS Induction of the pro-remodeling cytokine VEGF may be a potential mechanism by which UPM influences lung function growth in children irrespective of asthma status. Respiratory morbidity associated with UPM exposure in children may be related to increased expression of MUC5AC and IL-8.
Collapse
Affiliation(s)
- Kensho Iwanaga
- Division of Pediatric Pulmonary Medicine, Department of Pediatrics, University of California, San Francisco School of Medicine , San Francisco, CA , USA
| | | | | | | |
Collapse
|
30
|
A comprehensive analysis of oxidative stress in the ozone-induced lung inflammation mouse model. Clin Sci (Lond) 2014; 126:425-40. [PMID: 24040961 DOI: 10.1042/cs20130039] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Ozone is an oxidizing environmental pollutant that contributes significantly to respiratory health. Exposure to increased levels of ozone has been associated with worsening of symptoms of patients with asthma and COPD (chronic obstructive pulmonary disease). In the present study, we investigated the acute and chronic effects of ozone exposure-induced oxidative stress-related inflammation mechanics in mouse lung. In particular, we investigated the oxidative stress-induced effects on HDAC2 (histone deacetylase 2) modification and activation of the Nrf2 (nuclear factor erythroid-related factor 2) and HIF-1α (hypoxia-inducible factor-1α) signalling pathways. Male C57BL/6 mice were exposed to ozone (3 p.p.m.) for 3 h a day, twice a week for a period of 1, 3 or 6 weeks. Control mice were exposed to normal air. After the last exposure, mice were killed for BAL (bronchoalveolar lavage) fluid and lung tissue collection. BAL total cell counts were elevated at all of the time points studied. This was associated with increased levels of chemokines and cytokines in all ozone-exposed groups, indicating the presence of a persistent inflammatory environment in the lung. Increased inflammation and Lm (mean linear intercept) scores were observed in chronic exposed mice, indicating emphysematous changes were present in lungs of chronic exposed mice. The antioxidative stress response was active (indicated by increased Nrf2 activity and protein) after 1 week of ozone exposure, but this ability was lost after 3 and 6 weeks of ozone exposure. The transcription factor HIF-1α was elevated in 3- and 6-week ozone-exposed mice and this was associated with increased gene expression levels of several HIF-1α target genes including Hdac2 (histone deacetylase 2), Vegf (vascular endothelial growth factor), Keap1 (kelch-like ECH-associated protein 1) and Mif (macrophage migration inhibitory factor). HDAC2 protein was found to be phosphorylated and carbonylated in nuclear and cytoplasm fractions, respectively, and was associated with a decrease in DNA-binding activity and protein expression of HDAC2. Decreased HDAC2 activity, most likely a direct result of protein modification, in combination with the loss of the antioxidative stress response and activation of the HIF-1α pathway, contribute to the inflammatory response and emphysema observed in ozone-exposed mice.
Collapse
|
31
|
Yang B, Huang J, Xiang T, Yin X, Luo X, Huang J, Luo F, Li H, Li H, Ren G. Chrysin inhibits metastatic potential of human triple-negative breast cancer cells by modulating matrix metalloproteinase-10, epithelial to mesenchymal transition, and PI3K/Akt signaling pathway. J Appl Toxicol 2013; 34:105-12. [PMID: 24122885 DOI: 10.1002/jat.2941] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 08/25/2013] [Accepted: 09/06/2013] [Indexed: 11/10/2022]
Abstract
Chrysin, a naturally occurring flavone, has been shown to inhibit cell proliferation and induce cell apoptosis in various cancers. However, the effect and mechanisms of chrysin on cancer metastasis are still enigmatic. In this study, metastatic triple-negative breast cancer (TNBC) cell lines were used to evaluate the antimetastatic activity of chrysin. The results showed that chrysin (5, 10 and 20 μM) significantly suppressed TNBC cell migration and invasion in a dose-dependent manner. Human matrix metalloproteinase (MMP) antibody array demonstrated that MMP-10 was downregulated by chrysin, which was further verified by Western blotting and ELISA. Moreover, it was shown that chrysin induced increased E-cadherin expression and decreased expression of vimentin, snail and slug in TNBC cells, suggesting that chrysin had a reversal effect on epithelial-mesenchymal transition. More importantly, it was demonstrated that inhibiting the Akt signal pathway might play a central role in chrysin-induced antimetastatic activity by regulating MMP-10 and epithelial-mesenchymal transition. In conclusion, our study indicates that chrysin exerts antimetastatic activities in TNBC cells, which suggests that chrysin might be a potential therapeutic candidate for the treatment of advanced or metastatic breast cancer.
Collapse
Affiliation(s)
- Bing Yang
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bloom AJ, Baker TB, Chen LS, Breslau N, Hatsukami D, Bierut LJ, Goate A. Variants in two adjacent genes, EGLN2 and CYP2A6, influence smoking behavior related to disease risk via different mechanisms. Hum Mol Genet 2013; 23:555-61. [PMID: 24045616 DOI: 10.1093/hmg/ddt432] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Genome-wide significant associations with cigarettes per day (CPD) and risk for lung cancer and chronic obstructive pulmonary disease (COPD) were previously reported in a region of 19q13, including CYP2A6 (nicotine metabolism enzyme) and EGLN2 (hypoxia response). The associated single nucleotide polymorphisms (SNPs) were assumed to be proxies for functional variation in CYP2A6. Here, we demonstrate that when CYP2A6 and EGLN2 genotypes are analyzed together, the key EGLN2 variant, rs3733829, is not associated with nicotine metabolism independent of CYP2A6, but is nevertheless independently associated with CPD, and with breath carbon monoxide (CO), a phenotype associated with cigarette consumption and relevant to hypoxia. SNPs in EGLN2 are also associated with nicotine dependence and with smoking efficiency (CO/CPD). These results indicate a previously unappreciated novel mechanism behind genome-wide significant associations with cigarette consumption and disease risk unrelated to nicotine metabolism.
Collapse
Affiliation(s)
- A Joseph Bloom
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Susceptibility to viral infections in chronic obstructive pulmonary disease: role of epithelial cells. Curr Opin Pulm Med 2013; 19:125-32. [PMID: 23361194 DOI: 10.1097/mcp.0b013e32835cef10] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW The aim is to understand how airway epithelial cells with compromised innate defense mechanisms enhance susceptibility to respiratory virus infections in chronic obstructive pulmonary disease (COPD). RECENT FINDINGS Exacerbations associated with respiratory viruses are more severe and increase disease severity in COPD. Airway epithelial cells cultured from COPD patients show excessive innate immune response to viral infection and higher viral load compared with normal cells. SUMMARY Airway epithelial cells are the first line of defense in the lung and are equipped with several lines of innate defense mechanisms to fight against invading pathogens including viruses. Under normal conditions, mucociliary and barrier functions of airway epithelial cells prevent virus binding and entry into the cells. Virus-infected airway epithelial cells also express various cytokines, which recruit and activate innate and adaptive immune cells ultimately controlling the infection and tissue damage. In COPD however, compromised mucociliary and barrier functions may increase virus binding and allow virus entry into airway epithelial cells. Virus-infected COPD airway epithelial cells also show disproportionate cytokine expression leading to inappropriate recruitment and activation of innate and adaptive immune cells. COPD airway epithelial cells also show defective antiviral responses. Such defects in innate defense mechanisms may increase susceptibility to viral infections and disease severity in COPD.
Collapse
|
34
|
Yang J, Yu H, Zhou X, Kolosov V, Perelman J. Study on TRPV1-mediated mechanism for the hypersecretion of mucus in respiratory inflammation. Mol Immunol 2013; 53:161-71. [DOI: 10.1016/j.molimm.2012.06.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/20/2012] [Accepted: 06/24/2012] [Indexed: 02/06/2023]
|
35
|
Ganesan S, Unger BL, Comstock AT, Angel KA, Mancuso P, Martinez FJ, Sajjan US. Aberrantly activated EGFR contributes to enhanced IL-8 expression in COPD airways epithelial cells via regulation of nuclear FoxO3A. Thorax 2012; 68:131-41. [PMID: 23099361 DOI: 10.1136/thoraxjnl-2012-201719] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Decreased activity of forkhead transcription factor class O (FoxO)3A, a negative regulator of NF-κB-mediated chemokine expression, is implicated in the pathogenesis of chronic obstructive pulmonary disease (COPD). Previously, we showed that quercetin reduces lung inflammation in a murine model of COPD. Here, we examined the mechanisms underlying decreased FoxO3A activation and its modulation by quercetin in COPD human airway epithelial cells and in a COPD mouse model. METHODS Primary COPD and normal human airway epithelial cells were treated with quercetin, LY294002 or erlotinib for 2 weeks. IL-8 was measured by ELISA. FoxO3A, Akt, and epidermal growth factor (EGF) receptor (EGFR) phosphorylation and nuclear FoxO3A levels were determined by Western blot analysis. Effects of quercetin on lung chemokine expression, nuclear FoxO3A levels and phosphorylation of EGFR and Akt were determined in COPD mouse model. RESULTS Compared with normal, COPD cells showed significantly increased IL-8, which negatively correlated with nuclear FoxO3A levels. COPD bronchial biopsies also showed reduced nuclear FoxO3A. Decreased FoxO3A in COPD cells was associated with increased phosphorylation of EGFR, Akt and FoxO3A and treatment with quercetin, LY294002 or erlotinib increased nuclear FoxO3A and decreased IL-8 and phosphorylation of Akt, EGFR and FoxO3A, Compared with control, elastase/LPS-exposed mice showed decreased nuclear FoxO3A, increased chemokines and phosphorylation of EGFR and Akt. Treatment with quercetin partially reversed these changes. CONCLUSIONS In COPD airways, aberrant EGFR activity increases PI 3-kinase/Akt-mediated phosphorylation of FoxO3A, thereby decreasing nuclear FoxO3A and increasing chemokine expression. Quercetin restores nuclear FoxO3A and reduces chemokine expression partly by modulating EGFR/PI 3-kinase/Akt activity.
Collapse
Affiliation(s)
- Shyamala Ganesan
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109-5688, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Yang J, Zhang H, Zhao L, Chen Y, Liu H, Zhang T. Human adipose tissue-derived stem cells protect impaired cardiomyocytes from hypoxia/reoxygenation injury through hypoxia-induced paracrine mechanism. Cell Biochem Funct 2012; 30:505-14. [PMID: 22610511 DOI: 10.1002/cbf.2829] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 02/03/2012] [Accepted: 03/14/2012] [Indexed: 01/01/2023]
Affiliation(s)
- Junjie Yang
- Department of Cardiology; Chinese PLA General Hospital; Beijing; China
| | - Huaxin Zhang
- Department of Clinical Laboratory, Division of South Building; Chinese PLA General Hospital; Beijing; China
| | - Liang Zhao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center; Beijing Institute of Radiation Medicine; Beijing; China
| | - Yundai Chen
- Department of Cardiology; Chinese PLA General Hospital; Beijing; China
| | - Hongbin Liu
- Department of Cardiology; Chinese PLA General Hospital; Beijing; China
| | - Tao Zhang
- Department of Cardiology; Chinese PLA General Hospital; Beijing; China
| |
Collapse
|
37
|
Jang S, Park JW, Cha HR, Jung SY, Lee JE, Jung SS, Kim JO, Kim SY, Lee CS, Park HS. Silver nanoparticles modify VEGF signaling pathway and mucus hypersecretion in allergic airway inflammation. Int J Nanomedicine 2012; 7:1329-43. [PMID: 22457593 PMCID: PMC3310409 DOI: 10.2147/ijn.s27159] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The anti-inflammatory action of silver nanoparticles (NPs) has been reported in a murine model of asthma in a previous study. But more specific mechanisms of silver NPs in an attenuation of allergic airway inflammation have not yet been established. Vascular and mucous changes are believed to contribute largely in pathophysiology in asthma. Among various factors related to vascular changes, vascular endothelial growth factor (VEGF) plays a pivotal role in vascular changes in asthma. Mucin proteins MUC5AC and MUC5B have been implicated as markers of goblet cell metaplasia in lung pathologies. The aim of this study was to investigate the effects of silver NPs on VEGF signaling pathways and mucus hypersecretion. Ovalbumin (OVA)-inhaled female BALBc mice were used to evaluate the role of silver NPs and the related molecular mechanisms in allergic airway disease. In this study, with an OVA-induced murine model of allergic airway disease, it was found that the increased levels of hypoxia-inducible factor (HIF)-1α, VEGF, phosphatidylinositol-3 kinase (PI3K) and phosphorylated-Akt levels, and mucous glycoprotein expression (Muc5ac) in lung tissues were substantially decreased by the administration of silver NPs. In summary, silver NPs substantially suppressed mucus hypersecretion and PI3K/HIF-1α/VEGF signaling pathway in an allergic airway inflammation.
Collapse
Affiliation(s)
- Sunhyae Jang
- Division of Pulmonology, Allergy and Critical Care Medicine, Department of Internal Medicine, Chungnam National University Medical School, Daesadong, Daejeon, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|