1
|
Cheng ZJ, Luo YF, Zhu QY, Wang YF, Ren WY, Deng FY, Bo L, Jiang XY, Lei SF, Wu LF. Association of habitual glucosamine use with risk of microvascular complications among individuals with type 2 diabetes: a prospective cohort study in UK biobank. Nutr Diabetes 2025; 15:12. [PMID: 40169583 PMCID: PMC11961739 DOI: 10.1038/s41387-025-00369-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Glucosamine is a widely used supplement for treating osteoarthritis and joint pain. New evidence suggests a potential association between glucosamine and type 2 diabetes, inflammation and cardiometabolic risk. We aimed to prospectively evaluate the association of habitual glucosamine use with risk of diabetic microvascular complications based on data from the large-scale nationwide prospective UK Biobank cohort study. METHODS This analysis included 21,171 participants with type 2 diabetes who were free of microvascular complications from the UK Biobank. Incidence of diabetic microvascular complications was ascertained via electronic health records. The Cox proportional hazards model was used to assess the relationship between glucosamine use and the risk of diabetic microvascular complications. Subgroup analyses and sensitivity analyses were performed to explore the potential effect modifications and the robustness of the main findings. RESULTS At baseline, 14.5% of the participants reported habitual use of glucosamine supplements. During a median follow-up of 12.3 years, 4399 people developed diabetic microvascular complications, including 2084 cases of incident diabetic nephropathy, 2401 incident diabetic retinopathy, and 831 incident diabetic neuropathy. Glucosamine use was significantly associated with lower risks of composite microvascular complications (hazard ratio (HR) 0.89, 95% CI: 0.81 to 0.97) and diabetic nephropathy (HR 0.87, 95% CI: 0.76 to 0.98) in fully adjusted models. However, there was no significant inverse association between glucosamine use and the risk of diabetic retinopathy (HR 0.94, 95% CI: 0.83 to 1.06) or diabetic neuropathy (HR 0.88, 95% CI: 0.71 to 1.08). CONCLUSIONS Habitual use of glucosamine supplement was significantly associated with lower risks of composite microvascular complications and diabetic nephropathy but not retinopathy or neuropathy in individuals with type 2 diabetes.
Collapse
Affiliation(s)
- Zi-Jian Cheng
- Center for Genetic Epidemiology and Genomics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yu-Feng Luo
- Center for Genetic Epidemiology and Genomics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| | - Qing-Yun Zhu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| | - Yan-Fei Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| | - Wen-Yan Ren
- Cambridge-Suda Genomic Resource Center, Jiangsu Key Laboratory of Neuropsychiatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Fei-Yan Deng
- Center for Genetic Epidemiology and Genomics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China
| | - Lin Bo
- Department of Rheumatology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xi-Yuan Jiang
- Center of Osteoporosis, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China.
| | - Shu-Feng Lei
- Center for Genetic Epidemiology and Genomics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China.
| | - Long-Fei Wu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
2
|
Ge F, Sun Y, Han CC, Wei ZL, Guan X, Guo SW, Quan S, Zhou JG, Pang RP. Plasma Glutaminyl-Peptide Cyclotransferase Mediates Glucosamine-Metabolism-Driven Protection Against Hypertension: A Mendelian Randomization Study. Int J Mol Sci 2024; 25:12106. [PMID: 39596173 PMCID: PMC11593689 DOI: 10.3390/ijms252212106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Hypertension is one of the major risk factors for morbidity and mortality worldwide. In this study, Mendelian randomization was utilized to investigate how dietary supplement intake can impact hypertension based on circulating plasma metabolite genome-wide association study (GWAS) datasets, protein quantitative trait loci (pQTLs) of plasma proteins, and multiple public summary-level GWAS data. Pathway enrichment analysis combined with the results of inverse variance weighted Mendelian randomization revealed that a lower risk of hypertension was associated with the dietary intake of glucosamine, an anti-inflammatory supplement: odds ratio (OR) (95% CI): 0.888 (0.824-0.958). Additionally, glucosamine 6-phosphate N-acetyltransferase was identified as a protective factor against hypertension, OR (95% CI): 0.995 (0.992-0.998), shedding light on the potential protective mechanism of glucosamine. Mediation Mendelian randomization indicated that the protective effect of glucosamine metabolism was mediated by glutaminyl-peptide cyclotransferase, with a mediation proportion of 12.1% (5.9-18.2%), p < 0.05. This study offers new insights into preventive strategies for individuals with hypertension risk.
Collapse
Affiliation(s)
- Fei Ge
- Department of Pharmacology, Cardiac and Cerebrovascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yu Sun
- Department of Pharmacology, Cardiac and Cerebrovascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| | - Cong-Cong Han
- Department of Pharmacology, Cardiac and Cerebrovascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| | - Zi-Liang Wei
- Department of Pharmacology, Cardiac and Cerebrovascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xin Guan
- Department of Pharmacology, Cardiac and Cerebrovascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| | - Si-Wan Guo
- Department of Pharmacology, Cardiac and Cerebrovascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| | - Shui Quan
- Department of Pharmacology, Cardiac and Cerebrovascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| | - Jia-Guo Zhou
- Department of Pharmacology, Cardiac and Cerebrovascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| | - Rui-Ping Pang
- Department of Physiology, Pain Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
3
|
Meng M, Wu Y, Sha W, Zeng R, Luo D, Jiang R, Wu H, Zhuo Z, Yang Q, Li J, Leung FW, Duan C, Feng Y, Chen H. Associations of habitual glucosamine use with SARS-CoV-2 infection and hospital admission and death with COVID-19: Evidence from a large population based cohort study. J Med Virol 2023; 95:e28720. [PMID: 37185863 DOI: 10.1002/jmv.28720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has led to a fundamental number of morbidity and mortality worldwide. Glucosamine was indicated to help prevent and control RNA virus infection preclinically, while its potential therapeutic effects on COVID-19-related outcomes are largely unknown. To assess the association of habitual glucosamine use with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, hospital admission, and mortality with COVID-19 in a large population based cohort. Participants from UK Biobank were reinvited between June and September 2021 to have SARS-CoV-2 antibody testing. The associations between glucosamine use and the risk of SARS-CoV-2 infection were estimated by logistic regression. Hazard ratios (HRs) and 95% confidence intervals (CIs) for COVID-19-related outcomes were calculated using COX proportional hazards model. Furthermore, we carried out propensity-score matching (PSM) and stratified analyses. At baseline, 42 673 (20.7%) of the 205 704 participants reported as habitual glucosamine users. During median follow-up of 1.67 years, there were 15 299 cases of SARS-CoV-2 infection, 4214 cases of COVID-19 hospital admission, and 1141 cases of COVID-19 mortality. The fully adjusted odds ratio of SARS-CoV-2 infection with glucosamine use was 0.96 (95% CI: 0.92-1.01). The fully adjusted HR were 0.80 (95% CI: 0.74-0.87) for hospital admission, and 0.81 (95% CI: 0.69-0.95) for mortality. The logistic regression and Cox proportional hazard analyses after PSM yielded consistent results. Our study demonstrated that habitual glucosamine use is associated with reduced risks of hospital admission and death with COVID-19, but not the incidence of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Meijun Meng
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yanjun Wu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- Shantou University Medical College, Guangdong, China
| | - Ruijie Zeng
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Guangdong, China
| | - Dongling Luo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Rui Jiang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Huihuan Wu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Zewei Zhuo
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qi Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jingwei Li
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Guangdong, China
| | - Felix W Leung
- David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
- Sepulveda Ambulatory Care Center, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Chongyang Duan
- Department of Biostatistics, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yuliang Feng
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- Shantou University Medical College, Guangdong, China
| |
Collapse
|
4
|
Chen S, Wu H, Chen C, Wang D, Yang Y, Zhou Z, Zhu R, He X, Pan Y, Li C. The prognostic prediction of periodontal non-surgery therapy in periodontitis patients based on surface-enhanced Raman measurements of pre-treatment saliva. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 288:122150. [PMID: 36459721 DOI: 10.1016/j.saa.2022.122150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/15/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023]
Abstract
Periodontitis is one of the most prevalent dental diseases, and the patients with periodontitis often suffer from refractory periodontitis or recurrence of disease due to improper or inadequate treatment. In clinical practice, the early and accurate assessment of post-treatment prognosis in periodontitis patients is always very important in order to implement timely interventions. In this study, a pre-treatment saliva SERS based prognostic protocol was explored to predict the prognosis of periodontal non-surgery therapy in periodontitis patients. According to the biomolecular analysis, significant differences in the levels of ascorbic acid, uric acid and glutathione are observed between good prognosis group and poor prognosis group, which are expected to serve as potential prognostic markers. Furthermore, high accuracy, sensitivity and specificity can also be achieved by using the proposed prognostic model. The excellent performance of the proposed method has demonstrated its potential for fast, accurate, and non-invasive prognostic prediction of periodontal non-surgery therapy in periodontitis patients, even at the time before implementing treatment, thus is expected to benefit timely and rational guidance on clinical interventions.
Collapse
Affiliation(s)
- Shuo Chen
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China; Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, China
| | - Haotian Wu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Chen Chen
- School and Hospital of Stomatology, China Medical University, Shenyang, China; Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China
| | - Daheng Wang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Yaru Yang
- School and Hospital of Stomatology, China Medical University, Shenyang, China; Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China
| | - Zheng Zhou
- School of Innovation and Entrepreneurship, Liaoning Institute of Science and Technology, Benxi, China
| | - Ruochen Zhu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Xiaoning He
- The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yaping Pan
- School and Hospital of Stomatology, China Medical University, Shenyang, China; Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China
| | - Chen Li
- School and Hospital of Stomatology, China Medical University, Shenyang, China; Liaoning Provincial Key Laboratory of Oral Disease, Shenyang, China.
| |
Collapse
|
5
|
The role of transforming growth factor-β2 in cigarette smoke-induced lung inflammation and injury. Life Sci 2023; 320:121539. [PMID: 36870385 DOI: 10.1016/j.lfs.2023.121539] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 02/21/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023]
Abstract
AIMS Transforming growth factor-β2 (TGF-β2) plays an important role in pleiotropic functions and has been reported to be involved in the pathogenesis of chronic obstructive lung disease. The role of TGF-β2 in regulating cigarette smoke (CS)-induced lung inflammation and injury has not been investigated, and its underlying mechanism remains unclear. MAIN METHODS Primary bronchial epithelial cells (PBECs) were treated with cigarette smoke extract (CSE), and the signaling pathway of TGF-β2 regulating lung inflammation was investigated. Mice were exposed to CS and treated with TGF-β2 i.p. or bovine whey protein extract containing TGF-β2 p.o., and the role of TGF-β2 in alleviating lung inflammation/injury was studied. KEY FINDINGS In vitro, we demonstrated that TGF-β2 attenuated CSE-induced IL-8 production from PBECs through the TGF-β receptor I (TGF-βRI), Smad3, and mitogen-activated protein kinase signaling pathways. Selective TGF-βRI inhibitor (LY364947) and antagonist of Smad3 (SIS3) abolished the effect of TGF-β2 on alleviating CSE-induced IL-8 production. In vivo, CS exposure for 4 weeks in mice increased the levels of total protein, inflammatory cell counts, and monocyte chemoattractant protein-1 in bronchoalveolar fluid and induced lung inflammation/injury, as revealed by immunohistochemistry. Administration of TGF-β2 through intraperitoneal injection or oral feeding with bovine whey protein extract containing TGF-β2 significantly reduced CS-induced lung inflammation and injury. SIGNIFICANCE We concluded that TGF-β2 reduced CSE-induced IL-8 production through the Smad3 signaling pathway in PBECs and alleviated lung inflammation/injury in CS-exposed mice. The anti-inflammatory effect of TGF-β2 on CS-induced lung inflammation in humans deserves further clinical study.
Collapse
|
6
|
Spahr A, Divnic‐Resnik T. Impact of health and lifestyle food supplements on periodontal tissues and health. Periodontol 2000 2022; 90:146-175. [PMID: 35916868 PMCID: PMC9804634 DOI: 10.1111/prd.12455] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
According to the new classification, periodontitis is defined as a chronic multifactorial inflammatory disease associated with dysbiotic biofilms and characterized by progressive destruction of the tooth-supporting apparatus. This definition, based on the current scientific evidence, clearly indicates and emphasizes, beside the microbial component dental biofilm, the importance of the inflammatory reaction in the progressive destruction of periodontal tissues. The idea to modulate this inflammatory reaction in order to decrease or even cease the progressive destruction was, therefore, a logical consequence. Attempts to achieve this goal involve various kinds of anti-inflammatory drugs or medications. However, there is also an increasing effort in using food supplements or so-called natural food ingredients to modulate patients' immune responses and maybe even improve the healing of periodontal tissues. The aim of this chapter of Periodontology 2000 is to review the evidence of various food supplements and ingredients regarding their possible effects on periodontal inflammation and wound healing. This review may help researchers and clinicians to evaluate the current evidence and to stimulate further research in this area.
Collapse
Affiliation(s)
- Axel Spahr
- Discipline of Periodontics, School of Dentistry, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| | - Tihana Divnic‐Resnik
- Discipline of Periodontics, School of Dentistry, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
7
|
Zhan Y, Chen J, Wu J, Gu Y, Huang Q, Deng Z, Chen S, Wu X, Lv Y, Zeng Z, Xie J. Human epididymis protein 4 aggravates airway inflammation and remodeling in chronic obstructive pulmonary disease. Respir Res 2022; 23:120. [PMID: 35550579 PMCID: PMC9097053 DOI: 10.1186/s12931-022-02040-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/25/2022] [Indexed: 11/10/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a progressive disease characterized by chronic inflammation and airway remodeling. Human epididymis protein 4 (HE4) plays a critical role in various inflammatory or fibrotic diseases. However, the role of HE4 in COPD remains unidentified. Methods HE4 expression was determined in the lung tissues from COPD patients and cigarette smoke (CS)-exposed mice using immunohistochemical staining, qPCR, or western blot. The plasma level of HE4 was detected by ELISA. The regulations of HE4 in the expressions of CS extract (CSE)-induced inflammatory cytokines in human bronchial epithelial cells (HBE) were investigated through knockdown or overexpression of HE4. The role of secretory HE4 (sHE4) in the differentiation and proliferation in human pulmonary fibroblast cells (HPF) was explored via qPCR, western blot, CCK8 assay or 5-ethynyl-2′-deoxyuridine (EdU) staining. The probe of related mechanism in CSE-induced HE4 increase in HBE was conducted by administrating N-acetylcysteine (NAC). Results HE4 was up-regulated in both the lung tissue and plasma of COPD patients relative to controls, and the plasma HE4 was negatively associated with lung function in COPD patients. The same enhanced HE4 expression was verified in CS-exposed mice and CSE-induced HBE, but CSE failed to increase HE4 expression in HPF. In vitro experiments showed that reducing HE4 expression in HBE alleviated CSE-induced IL-6 release while overexpressing HE4 facilitated IL-6 expression, mechanistically through affecting phosphorylation of NFκB-p65, whereas intervening HE4 expression had no distinctive influence on IL-8 secretion. Furthermore, we confirmed that sHE4 promoted fibroblast-myofibroblast transition, as indicated by promoting the expression of fibronectin, collagen I and α-SMA via phosphorylation of Smad2. EdU staining and CCK-8 assay demonstrated the pro-proliferative role of sHE4 in HPF, which was further confirmed by enhanced expression of survivin and PCNA. Pretreatment of NAC in CSE or H2O2-induced HBE mitigated HE4 expression. Conclusions Our study indicates that HE4 may participate in airway inflammation and remodeling of COPD. Cigarette smoke enhances HE4 expression and secretion in bronchial epithelium mediated by oxidative stress. Increased HE4 promotes IL-6 release in HBE via phosphorylation of NFκB-p65, and sHE4 promotes fibroblastic differentiation and proliferation. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02040-7.
Collapse
Affiliation(s)
- Yuan Zhan
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430030, Hubei, China
| | - Jinkun Chen
- Department of Science, Western University, 1151 Richmond Street, London, ON, N6A 3K7, Canada
| | - Jixing Wu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430030, Hubei, China
| | - Yiya Gu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430030, Hubei, China
| | - Qian Huang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430030, Hubei, China
| | - Zhesong Deng
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430030, Hubei, China
| | - Shanshan Chen
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430030, Hubei, China
| | - Xiaojie Wu
- Department of Respiratory and Critical Care Medicine, Wuhan NO.1 Hospital, Wuhan Hospital of Traditional Chinese and Western Medicine, Wuhan, 430022, China
| | - Yongman Lv
- Health Management Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Zhilin Zeng
- Department and Institute of Infectious Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, China.
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Wuhan, 430030, Hubei, China.
| |
Collapse
|
8
|
Cipollina C, Bruno A, Fasola S, Cristaldi M, Patella B, Inguanta R, Vilasi A, Aiello G, La Grutta S, Torino C, Pace E. Cellular and Molecular Signatures of Oxidative Stress in Bronchial Epithelial Cell Models Injured by Cigarette Smoke Extract. Int J Mol Sci 2022; 23:1770. [PMID: 35163691 PMCID: PMC8836577 DOI: 10.3390/ijms23031770] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 02/06/2023] Open
Abstract
Exposure of the airways epithelium to environmental insults, including cigarette smoke, results in increased oxidative stress due to unbalance between oxidants and antioxidants in favor of oxidants. Oxidative stress is a feature of inflammation and promotes the progression of chronic lung diseases, including Chronic Obstructive Pulmonary Disease (COPD). Increased oxidative stress leads to exhaustion of antioxidant defenses, alterations in autophagy/mitophagy and cell survival regulatory mechanisms, thus promoting cell senescence. All these events are amplified by the increase of inflammation driven by oxidative stress. Several models of bronchial epithelial cells are used to study the molecular mechanisms and the cellular functions altered by cigarette smoke extract (CSE) exposure, and to test the efficacy of molecules with antioxidant properties. This review offers a comprehensive synthesis of human in-vitro and ex-vivo studies published from 2011 to 2021 describing the molecular and cellular mechanisms evoked by CSE exposure in bronchial epithelial cells, the most used experimental models and the mechanisms of action of cellular antioxidants systems as well as natural and synthetic antioxidant compounds.
Collapse
Affiliation(s)
- Chiara Cipollina
- Ri.MED Foundation, 90133 Palermo, Italy; (C.C.); (M.C.)
- Institute for Biomedical Research and Innovation, National Research Council, 90146 Palermo, Italy; (A.B.); (S.F.); (S.L.G.); (E.P.)
| | - Andreina Bruno
- Institute for Biomedical Research and Innovation, National Research Council, 90146 Palermo, Italy; (A.B.); (S.F.); (S.L.G.); (E.P.)
- Institute of Translational Pharmacology, National Research Council, 90146 Palermo, Italy
| | - Salvatore Fasola
- Institute for Biomedical Research and Innovation, National Research Council, 90146 Palermo, Italy; (A.B.); (S.F.); (S.L.G.); (E.P.)
- Institute of Translational Pharmacology, National Research Council, 90146 Palermo, Italy
| | | | - Bernardo Patella
- Department of Engineering, University of Palermo, 90128 Palermo, Italy; (B.P.); (R.I.); (G.A.)
| | - Rosalinda Inguanta
- Department of Engineering, University of Palermo, 90128 Palermo, Italy; (B.P.); (R.I.); (G.A.)
| | - Antonio Vilasi
- Institute of Clinical Physiology, National Research Council, 89124 Reggio Calabria, Italy;
| | - Giuseppe Aiello
- Department of Engineering, University of Palermo, 90128 Palermo, Italy; (B.P.); (R.I.); (G.A.)
| | - Stefania La Grutta
- Institute for Biomedical Research and Innovation, National Research Council, 90146 Palermo, Italy; (A.B.); (S.F.); (S.L.G.); (E.P.)
- Institute of Translational Pharmacology, National Research Council, 90146 Palermo, Italy
| | - Claudia Torino
- Institute of Clinical Physiology, National Research Council, 89124 Reggio Calabria, Italy;
| | - Elisabetta Pace
- Institute for Biomedical Research and Innovation, National Research Council, 90146 Palermo, Italy; (A.B.); (S.F.); (S.L.G.); (E.P.)
- Institute of Translational Pharmacology, National Research Council, 90146 Palermo, Italy
| |
Collapse
|
9
|
Cui M, Wang Y, Liu K. Structure analysis and anti-inflammatory effect of a polysaccharide from Hippocampus mohnikei. Chem Biodivers 2022; 19:e202100739. [PMID: 35001528 DOI: 10.1002/cbdv.202100739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 01/06/2022] [Indexed: 11/10/2022]
Abstract
The purpose of this study was to analyze the structure of a polysaccharide (HMP-1) from Hippocampus mohnikei, and to explore its anti-inflammatory effect. HMP-1 was obtained from Hippocampus mohnikei by ethanol sedimentation and secondary column chromatography purification. Its structural characteristics were analyzed by gel permeation chromatography (GPC), fourier transform infrared (FT-IR) spectroscopy, nuclear magnetic resonance (NMR) spectroscopy and scanning electron micrograph (SEM). Results showed its molecular weight (Mw) was 7296 Da, and it mainly consisted of six residues, 1,3-β-Glcp, 1,4-α-Manp, 1,4-α-GalpA, 1,4-β-GlcpA2S, 1,4-α-Galp3S, 1,4-β-GlcNAc. HMP-1 could protect RAW246.7 cells from the cytotoxic effect induced by LPS. HMP-1 also could reduce the levels of nitric oxide and reactive oxygen species produced by LPS stimulation, suggesting that HMP-1 has anti-inflammatory activities within a certain concentration range.
Collapse
Affiliation(s)
- Mingxiao Cui
- Shanghai Ocean University, College of food science and technology, 999 Hucheng Ring Road, Pudong New Area, 201306, Shanghai, CHINA
| | - Yu Wang
- Shanghai Ocean University, College of food science and technology, 999 Hucheng Ring Road, Pudong New Area, Shanghai, CHINA
| | - Kehai Liu
- Shanghai Ocean University, college of food science and technology, 999 Hucheng Ring Road, Pudong New Area, Shanghai, CHINA
| |
Collapse
|
10
|
Jung SY, Kim GD, Choi DW, Shin DU, Eom JE, Kim SY, Chai OH, Kim HJ, Lee SY, Shin HS. Epilobiumpyrricholophum Extract Suppresses Porcine Pancreatic Elastase and Cigarette Smoke Extract-Induced Inflammatory response in a Chronic Obstructive Pulmonary Disease Model. Foods 2021; 10:foods10122929. [PMID: 34945480 PMCID: PMC8700438 DOI: 10.3390/foods10122929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 01/01/2023] Open
Abstract
Chronic airway exposure to harmful substances, such as deleterious gases, cigarette smoke (CS), and particulate matter, triggers chronic obstructive pulmonary disease (COPD), characterized by impaired lung function and unbridled immune responses. Emerging epigenomic and genomic evidence suggests that excessive recruitment of alveolar macrophages and neutrophils contributes to COPD pathogenesis by producing various inflammatory mediators, such as reactive oxygen species (ROS), neutrophil elastase, interleukin (IL) 6, and IL8. Recent studies showed that Epilobium species attenuated ROS, myeloperoxidase, and inflammatory cytokine production in murine and human innate immune cells. Although the Epilobium genus exerts anti-inflammatory, antioxidant, and antimicrobial effects, the question of whether the Epilobium species regulate lung inflammation and innate immune response in COPD has not been investigated. In this study, Epilobium pyrricholophum extract (EPE) suppressed inflammatory cell recruitment and clinical symptoms in porcine pancreatic elastase and CS extract-induced COPD mice. In addition, EPE attenuated inflammatory gene expression by suppressing MAPKs and NFκB activity. Furthermore, UPLC-Q-TOF MS analyses revealed the anti-inflammatory effects of the identified phytochemical constituents of EPE. Collectively, our studies revealed that EPE represses the innate immune response and inflammatory gene expression in COPD pathogenesis in mice. These findings provide insights into new therapeutic approaches for treating COPD.
Collapse
Affiliation(s)
- Sun Young Jung
- Research Division of Food Functionality, Korea Food Research Institute, Wanju 55365, Korea; (S.Y.J.); (G.-D.K.); (D.W.C.)
- Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Korea;
| | - Gun-Dong Kim
- Research Division of Food Functionality, Korea Food Research Institute, Wanju 55365, Korea; (S.Y.J.); (G.-D.K.); (D.W.C.)
| | - Dae Woon Choi
- Research Division of Food Functionality, Korea Food Research Institute, Wanju 55365, Korea; (S.Y.J.); (G.-D.K.); (D.W.C.)
- Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Korea;
| | - Dong-Uk Shin
- Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Korea;
- Research Group of Natural Materials and Metabolism, Korea Food Research Institute, Wanju 55365, Korea
| | - Ji-Eun Eom
- Food Functional Evaluation Support Team, Korea Food Research Institute, Wanju 55365, Korea;
| | - Seung Yong Kim
- Department of Food Science and Technology, Jeonbuk National University, Jeonju 54896, Korea;
| | - Ok Hee Chai
- Department of Anatomy, Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea;
| | - Hyun-Jin Kim
- Division of Applied Life Science (BK21 Four), Department of Food Science and Technology, and Institute of Agriculture and Life Science, Gyeongsang National University, 501 Jinjudaero, Jinju 52828, Gyeongsangnam-do, Korea;
- EZmass. Co. Ltd., 501 Jinjudaero, Jinju 55365, Gyeongsangnam-do, Korea
| | - So-Young Lee
- Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Korea;
- Research Group of Natural Materials and Metabolism, Korea Food Research Institute, Wanju 55365, Korea
- Correspondence: (S.-Y.L.); (H.S.S.); Tel.: +82-63-219-9348 (S.-Y.L.); +82-63-219-9296 (H.S.S.)
| | - Hee Soon Shin
- Research Division of Food Functionality, Korea Food Research Institute, Wanju 55365, Korea; (S.Y.J.); (G.-D.K.); (D.W.C.)
- Department of Food Biotechnology, University of Science and Technology, Daejeon 34113, Korea;
- Correspondence: (S.-Y.L.); (H.S.S.); Tel.: +82-63-219-9348 (S.-Y.L.); +82-63-219-9296 (H.S.S.)
| |
Collapse
|
11
|
Huang Y, Zhou P, Liu S, Duan W, Zhang Q, Lu Y, Wei X. Metabolome and microbiome of chronic periapical periodontitis in permanent anterior teeth: a pilot study. BMC Oral Health 2021; 21:599. [PMID: 34814909 PMCID: PMC8609808 DOI: 10.1186/s12903-021-01972-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 11/26/2022] Open
Abstract
Background Periapical periodontitis is a common oral inflammatory disease that affects periapical tissues and is caused by bacteria in the root canal system. The relationship among the local metabolome, the inflammatory grade, and the type and abundance of microorganisms associated with periapical periodontitis is discussed in this study. Methods The inflammatory grades of periapical samples from 47 patients with chronic periapical periodontitis in permanent anterior teeth were determined based on the immune cell densities in tissues subjected to haematoxylin and eosin staining. The metabolome was evaluated using ultrahigh-performance liquid chromatography-quadrupole time-of-flight mass spectrometry, followed by principal component analysis and orthogonal partial least squares discriminant analysis. The microbiome was accessed using 16 S rRNA high-throughput sequencing. The differences in the metabolomes and microbiomes of the periapical periodontitis samples were assessed using Spearman’s correlation analysis. Result N-acetyl-D-glucosamine, L-tryptophan, L-phenylalanine, and 15 other metabolites were identified by the comparison between samples with severe inflammation and mild or moderate inflammation. Four amino acid metabolism pathways and one sugar metabolism pathway were associated with the inflammatory grade of periapical periodontitis. The abundance of Actinomycetes was negatively correlated with the abundance of glucosamine (GlcN), while the abundance of Tannerella was positively correlated with the abundance of L-methionine. Conclusions The local metabolome of periapical periodontitis is correlated with the inflammatory grade. The abundance of the local metabolites GlcN and L-methionine is correlated with the abundance of the major microorganisms Actinomycetes and Tannerella, respectively.
Collapse
Affiliation(s)
- Yun Huang
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Conservative Dentistry and Endodontics, Stomatological Hospital, Nanjing Medical University, Nanjing, China.,Department of Operative Dentistry and Endodontics, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Peng Zhou
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Conservative Dentistry and Endodontics, Stomatological Hospital, Nanjing Medical University, Nanjing, China.,Department of Operative Dentistry and Endodontics, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Siqi Liu
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Conservative Dentistry and Endodontics, Stomatological Hospital, Nanjing Medical University, Nanjing, China.,Department of Operative Dentistry and Endodontics, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Wei Duan
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Conservative Dentistry and Endodontics, Stomatological Hospital, Nanjing Medical University, Nanjing, China.,Department of Operative Dentistry and Endodontics, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Qinqin Zhang
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Conservative Dentistry and Endodontics, Stomatological Hospital, Nanjing Medical University, Nanjing, China.,Department of Operative Dentistry and Endodontics, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Ying Lu
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Conservative Dentistry and Endodontics, Stomatological Hospital, Nanjing Medical University, Nanjing, China.,Department of Operative Dentistry and Endodontics, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China
| | - Xin Wei
- Jiangsu Province Key Laboratory of Oral Diseases, Department of Conservative Dentistry and Endodontics, Stomatological Hospital, Nanjing Medical University, Nanjing, China. .,Department of Operative Dentistry and Endodontics, Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China. .,Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing, China.
| |
Collapse
|
12
|
Tian X, Xue Y, Xie G, Zhou Y, Xiao H, Ding F, Zhang M. (-)-Epicatechin ameliorates cigarette smoke-induced lung inflammation via inhibiting ROS/NLRP3 inflammasome pathway in rats with COPD. Toxicol Appl Pharmacol 2021; 429:115674. [PMID: 34403689 DOI: 10.1016/j.taap.2021.115674] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 11/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) with increased morbidity and mortality is a worldwide healthcare challenge closely associated with cigarette smoking (CS). Currently, there is no effective therapeutic strategy to control inflammation in COPD patients. The present study tested the protective effects of (-)-Epicatechin (EC), a type of flavonoid, on CS-induced COPD and the underlying mechanism. Also, EC repressed the production of reactive oxygen species (ROS) and improved human bronchial epithelial cell viability after cigarette smoke extract (CSE) treatment. Further studies demonstrated that EC promotes ubiquitin-mediated Keap1 degradation by upregulating tripartite motif-containing protein 25 (TRIM25) expression and enhances the nuclear localization of Nrf2 protein. Also, EC dramatically inhibits the activation of NLRP3 inflammasome and reduces the CSE-induced pyroptosis, as indicated by decreasing lactate dehydrogenase release and the number of caspase-1-positive cells. Importantly, Nrf2 knockdown reversed the protective effect of EC on human bronchial epithelial cells, at least partially. Consistent with the results in vitro, EC inhibits the activation of NLRP3 inflammasome and relieves the CS-induced lung inflammation, as evident from decreased interleukin (IL)-1β and IL-18 secretion in a COPD rat model. In conclusion, this study revealed the protective effect of EC on experimental COPD rats and elucidated the mechanism of EC promoting Nrf2 activity, which might provide a novel therapeutic strategy for COPD.
Collapse
Affiliation(s)
- Xue Tian
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine,100 Haining Road, Shanghai 200080, PR China
| | - Yishu Xue
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine,100 Haining Road, Shanghai 200080, PR China
| | - Guogang Xie
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine,100 Haining Road, Shanghai 200080, PR China
| | - Yan Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine,100 Haining Road, Shanghai 200080, PR China
| | - Hui Xiao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine,100 Haining Road, Shanghai 200080, PR China
| | - Fengming Ding
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine,100 Haining Road, Shanghai 200080, PR China
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine,100 Haining Road, Shanghai 200080, PR China..
| |
Collapse
|
13
|
Eurotium cristatum Fermented Loose Dark Tea Ameliorates Cigarette Smoke-Induced Lung Injury by MAPK Pathway and Enhances Hepatic Metabolic Detoxification by PXR/ AhR Pathway in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6635080. [PMID: 33777316 PMCID: PMC7972846 DOI: 10.1155/2021/6635080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/05/2021] [Accepted: 02/26/2021] [Indexed: 01/28/2023]
Abstract
Cigarette smoke- (CS-) induced oxidative stress and inflammation in the lung are serious health problems. Primary and reprocessed tea products contain multiple antioxidants that have been reported to protect the lung against CS-induced injury. However, the beneficial effects of Eurotium cristatum fermented loose dark tea (ECT) and Eurotium cristatum particle metabolites (ECP) on CS-induced lung injury and its potential hepatic metabolic detoxification are still unclear. Therefore, sixty mice were randomly divided into six equal groups. CS-exposed mice were prevented or treated with ECP or ECT infusions for 12 or 8 weeks to determine the antioxidative stress, anti-inflammatory and potential metabolic detoxification of ECT and ECP. Thirty-six mice were randomly divided into six equal groups to observe the effects on hepatic metabolic detoxification by replacing daily drinking water with ECT. Results showed that CS significantly decreased the activities of glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD) and upregulated the expressions of malondialdehyde (MDA), tumor necrosis factor alpha (TNF-α), interleukin-6 (IL-6), IL-8, and IL-1β in serum. These adverse effects were modulated by ECP and ECT. In addition, ECT upregulated the mRNA expression of pregnane X receptor (PXR) and cytochrome P450 (CYP450) in the liver on daily free drinking ECT mice group. Western blot analysis further revealed that in CS-exposed mice, ECP and ECT significantly decreased the phosphorylation of mitogen-activated protein kinase (MAPK) in the lung but upregulated the protein expressions of PXR and aryl hydrocarbon receptor (AhR) in the liver. Overall, our findings demonstrated that ECT and ECP protected against lung injury induced by CS via MAPK pathway and enhanced hepatic metabolic detoxification via PXR and AhR pathways. Therefore, daily intake of ECT and ECP can potentially protect against CS-induced oxidative and inflammatory injuries.
Collapse
|
14
|
DiNicolantonio JJ, Barroso-Aranda J. Harnessing adenosine A2A receptors as a strategy for suppressing the lung inflammation and thrombotic complications of COVID-19: Potential of pentoxifylline and dipyridamole. Med Hypotheses 2020; 143:110051. [PMID: 32650197 PMCID: PMC7330590 DOI: 10.1016/j.mehy.2020.110051] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 12/30/2022]
Abstract
Counterproductive lung inflammation and dysregulated thrombosis contribute importantly to the lethality of advanced COVID-19. Adenosine A2A receptors (A2AR), expressed by a wide range of immune cells, as well as endothelial cells and platelets, exert cAMP-mediated anti-inflammatory and anti-thrombotic effects that potentially could be highly protective in this regard. The venerable drug pentoxifylline (PTX) exerts both anti-inflammatory and antithrombotic effects that reflect its ability to boost the responsiveness of A2AR to extracellular adenosine. The platelet-stabilizing drug dipyridamole (DIP) blocks intracellular uptake of extracellularly-generated adenosine, thereby up-regulating A2AR signaling in a way that should be functionally complementary to the impact of PTX in that regard. Moreover, DIP has recently been reported to slow the cellular replication of SARS-CoV-2 in clinically feasible concentrations. Both PTX and DIP are reasonably safe, well-tolerated, widely available, and inexpensive drugs. When COVID-19 patients can be treated within several days of symptom onset, using PTX + DIP in conjunction with hydroxychloroquine (HCQ) and an antibiotic – azithromycin (AZM) or doxycycline – might be warranted. HCQ and AZM can suppress SARS-CoV-2 proliferation in vitro and may slow the cell-to-cell spread of the virus; a large case series evaluating this combination in early-stage patients reported an impressively low mortality rate. However, whereas HCQ and AZM can promote QT interval lengthening and may be contraindicated in more advanced COVID-19 entailing cardiac damage, doxycycline has no such effect and exerts a potentially beneficial anti-inflammatory action. In contrast to HCQ, we propose that the combination of PTX + DIP can be used in both early and advanced stages of COVID-19. Concurrent use of certain nutraceuticals – yeast beta-glucan, zinc, vitamin D, spirulina, phase 2 inducers, N-acetylcysteine, glucosamine, quercetin, and magnesium – might also improve therapeutic outcomes in COVID-19.
Collapse
|
15
|
Takano M, Higa S, Furuichi Y, Naka R, Yumoto R. Suppression of P-glycoprotein by cigarette smoke extract in human lung-derived A549/P-gp cells. Drug Metab Pharmacokinet 2020; 35:214-219. [PMID: 32037157 DOI: 10.1016/j.dmpk.2019.12.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 11/16/2022]
Abstract
Effect of long-term treatment with cigarette smoke extract (CSE) on the function and expression of P-glycoprotein (P-gp) in lung alveolar epithelial cells was examined using A549/P-gp cell line expressing P-gp. CSE treatment suppressed P-gp activity in a concentration- and treatment time-dependent manner. The suppression of P-gp activity by CSE was irreversible for at least 96 h after removal of CSE. In addition, CSE treatment suppressed the expression of P-gp mRNA and protein. In order to understand the mechanisms underlying P-gp suppression by CSE, the role of reactive oxygen species (ROS) was examined. CSE treatment increased intracellular ROS level, and suppressed catalase activity. α-Tocopherol suppressed ROS production by CSE, and ameliorated the suppression of P-gp activity by CSE, suggesting that ROS is involved in CSE-induced suppression of P-gp. The role of intracellular signaling pathways such as the nuclear factor κB and mitogen-activated protein kinase pathways was also examined. Among these pathways, the involvement of extracellular signal-regulated kinase (ERK) pathway was suggested. Taken together, long-term CSE treatment may suppress P-gp via modulation of ROS level and ERK pathway in alveolar epithelial cells.
Collapse
Affiliation(s)
- Mikihisa Takano
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Shuhei Higa
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Yuma Furuichi
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Ryosuke Naka
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Ryoko Yumoto
- Department of Pharmaceutics and Therapeutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| |
Collapse
|
16
|
Cheki M, Jafari S, Najafi M, Mahmoudzadeh A. Glucosamine Protects Rat Bone Marrow Cells Against Cisplatin-induced Genotoxicity and Cytotoxicity. Anticancer Agents Med Chem 2019; 19:1695-1702. [PMID: 31272360 DOI: 10.2174/1871520619666190704164126] [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] [Received: 04/05/2019] [Revised: 05/20/2019] [Accepted: 05/21/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND OBJECTIVE Glucosamine is a widely prescribed dietary supplement used in the treatment of osteoarthritis. In the present study, the chemoprotectant ability of glucosamine was evaluated against cisplatin-induced genotoxicity and cytotoxicity in rat bone marrow cells. METHODS Glucosamine was orally administrated to rats at doses of 75 and 150 mg/kg body weight for seven consecutive days. On the seventh day, the rats were treated with a single injection of cisplatin (5 mg/kg, i.p.) at 1h after the last oral administration. The cisplatin antagonistic potential of glucosamine was assessed by micronucleus assay, Reactive Oxygen Species (ROS) level analysis, hematological analysis, and flow cytometry. RESULTS Glucosamine administration to cisplatin-treated rats significantly decreased the frequencies of Micronucleated Polychromatic Erythrocytes (MnPCEs) and Micronucleated Normchromatic Erythrocytes (MnNCEs), and also increased PCE/(PCE+NCE) ratio in bone marrow cells. Furthermore, treatment of rats with glucosamine before cisplatin significantly inhibited apoptosis, necrosis and ROS generation in bone marrow cells, and also increased red blood cells count in peripheral blood. CONCLUSION This study shows glucosamine to be a new effective chemoprotector against cisplatin-induced DNA damage and apoptosis in rat bone marrow cells. The results of this study may be helpful in reducing the harmful effects of cisplatin-based chemotherapy in the future.
Collapse
Affiliation(s)
- Mohsen Cheki
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Salman Jafari
- Department of Radiology Technology, School of Paramedicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Aziz Mahmoudzadeh
- Department of Biosciences and Biotechnology, Malek-Ashtar University of Technology, Tehran, Iran
| |
Collapse
|
17
|
Li F, Song X, Su G, Wang Y, Wang Z, Qing S, Jia J, Wang Y, Huang L, Zheng K, Wang Y. AT-533, a Hsp90 inhibitor, attenuates HSV-1-induced inflammation. Biochem Pharmacol 2019; 166:82-92. [PMID: 31071330 DOI: 10.1016/j.bcp.2019.05.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/02/2019] [Indexed: 12/11/2022]
Abstract
Inflammatory events are tightly associated with the death caused by Herpes simplex virus 1 (HSV-1) infection of the brain. Heat shock protein 90 (Hsp90) is a molecular chaperone that is stimulated in response to many stressful conditions (e.g., inflammation and hypoxia) and Hsp90 inhibitors are suggested to be potent inhibitors of the inflammatory response. The aim of this study was to investigate the effect of Hsp90 inhibitor AT-533 on HSV-1-induced inflammation. AT-533 at a non-antiviral concentration was found to show a prominent inhibitory effect on the production of cytokines induced by HSV-1 infection, such as tumor necrosis factor α (TNF-α), interleukin 6 (IL-6) and interleukin 1β (IL-1β). Mechanically, HSV-1 early infection induced inflammation through NF-κB signaling and NLRP3 inflammasome activation, as illustrated by the nuclear translocation of NF-κB and the enhanced cleavage of caspase-1. Besides, HSV-1 enhanced the interaction between NLRP3 and Hsp90. Moreover, AT-533 reduced the nuclear translocation of NF-κB and inflammasome activation via inhibiting the chaperone function of Hsp90. Furthermore, AT-533 inhibited the cleavage of pro-IL-1β to mature IL-1β in a NLRP3-independent manner. In summary, AT-533 may be a promising therapeutic strategy in HSV-1-infected inflammation management.
Collapse
Affiliation(s)
- Feng Li
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xiaowei Song
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Guifeng Su
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yiliang Wang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zhaoyang Wang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Shurong Qing
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Jiaoyan Jia
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yuan Wang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Lianzhou Huang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Kai Zheng
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China.
| | - Yifei Wang
- Institute of Biomedicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
18
|
Glucosamine inhibits IL-1β expression by preserving mitochondrial integrity and disrupting assembly of the NLRP3 inflammasome. Sci Rep 2019; 9:5603. [PMID: 30944389 PMCID: PMC6447579 DOI: 10.1038/s41598-019-42130-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 03/21/2019] [Indexed: 12/18/2022] Open
Abstract
The NLRP3 inflammasome promotes the pathogenesis of metabolic, neurodegenerative and infectious diseases. Increasing evidences show that the NLRP3 inflammasome is a promising therapeutic target in inflammatory diseases. Glucosamine is widely used as a dietary supplement to promote the health of cartilage tissue and is expected to exert anti-inflammatory activity in joint inflammation, which is a nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome-associated complication. Here, we investigated whether GlcN inhibits the NLRP3 inflammasome and dissected the underlying molecular mechanisms. We found that GlcN suppressed the NLRP3 inflammasome in mouse and human macrophages. A mechanistic study revealed that GlcN inhibited the expression of NLRP3 and IL-1β precursor by reducing reactive oxygen species generation and NF-κB activation in lipopolysaccharide-activated macrophages. GlcN also suppressed mitochondrial reactive oxygen species generation and mitochondrial integrity loss in NLRP3-activated macrophages. Additionally, GlcN disrupted NLRP3 inflammasome assembly by inhibiting NLRP3 binding to PKR, NEK7 and ASC. Furthermore, oral administration of GlcN reduced peritoneal neutrophils influx and lavage fluids concentrations of IL-1β, IL-6 MCP-1 and TNF-α in uric acid crystal-injected mice. These results indicated that GlcN might be a novel dietary supplement for the amelioration of NLRP3 inflammasome-associated complications.
Collapse
|
19
|
Wang J, Yang G, Li M, Zhou X. Transient Receptor Potential Melastatin 8 (TRPM8)-Based Mechanisms Underlie Both the Cold Temperature-Induced Inflammatory Reactions and the Synergistic Effect of Cigarette Smoke in Human Bronchial Epithelial (16HBE) Cells. Front Physiol 2019; 10:285. [PMID: 31001124 PMCID: PMC6455074 DOI: 10.3389/fphys.2019.00285] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 03/04/2019] [Indexed: 12/25/2022] Open
Abstract
Transient receptor potential melastatin 8 (TRPM8) is a major receptor of cold environment. Recently, we found that cigarette smoke extract (CSE) upregulated TRPM8 mRNA and protein expression in bronchial tissues that made them more sensitive to cold stimuli. In our present study, we found that cold temperature (18°C)-induced activation of TRPM8 in 16HBE (human bronchial epithelial) cells facilitated Ca2+ influx and subsequently led to the increased expression of interleukin (IL)-6, IL-8, and tumor necrosis factor (TNF)-α via the upregulation of p-extracellular signal-regulated kinase (ERK) and the activation of NF-κB. In addition, 16HBE cells that co-stimulated with 18°C and CSE were used to explore the synergistic effect of CSE on cold temperature-induced inflammatory cytokine production as well as the possible involved signaling pathway. RT-PCR and western blot analysis revealed that CSE upregulated TRPM8 mRNA and protein level in 16HBE cells. Ca2+ imaging, western blot, and luciferase assay showed more robust increase in intracellular Ca2+ and promoted phosphorylated ERK, P38, and NF-κB activity, respectively, in 16HBE cells co-stimulated with CSE and cold temperature, and such alteration was attenuated by TRPM8 short hairpin RNA (shRNA) transfection and BCTC pretreatment. Furthermore, enhanced levels of IL-6, IL-8, and TNF-α showed by enzyme-linked immunosorbent assay (ELISA) were reduced by specific inhibitors of ERK and NF-κB. Collectively, our results suggest that mitogen-activated protein kinase (MAPK)/NF-κB signaling is involved in TRPM8-mediated cold temperature-induced inflammatory cytokine expression. In addition, CSE synergistically amplifies cold temperature-induced inflammatory factors release via upregulating TRPM8 expression and enhancing MAPK/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Jing Wang
- Department of Respiratory Medicine, The Second Clinical Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Yang
- Department of Neurosurgery, The First Clinical Hospital of Chongqing Medical University, Chongqing, China
| | - Minchao Li
- Department of Respiratory Medicine, The Second Clinical Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangdong Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
20
|
Basiri F, Rad A, Mahdian D, Molavi M, Amin B. Effects of glucosamine against morphine-induced antinociceptive tolerance and dependence in mice. J Biomed Sci 2019; 26:21. [PMID: 30782159 PMCID: PMC6380027 DOI: 10.1186/s12929-019-0513-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 02/11/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The most important limitations of morphine in pain therapy are its tolerance and dependence. In this study, we evaluated the protective effect of glucosamine against morphine-induced tolerance and dependence in mice. METHODS Mice received twice daily morphine (20 mg/kg, s.c.) alone, or along with orally administered glucosamine (500, 1000 and 2000 mg/kg), for 9 continuous days. To assess antinociceptive effect of morphine, percentage of maximal possible effect (%MPE) of animals exposed to thermal stimulus was measured in the hot plate test, 30 min after morphine administration. Test was performed on days 1, 3, 5, 7 and 9. The effect of glucosamine on the naloxone (5 mg/kg, i.p.)-precipitated morphine withdrawal, was also evaluated. Changes in brain gene expression levels of induced nitric oxide synthase (iNOS), enzyme responsible for nitric oxide generation, as well as pro-inflammatory mediator, tumor necrosis alpha (TNF-α) were measured in morphine tolerated animals, as well as after withdrawal by real-time polymerase chain reaction (RT-PCR). Protein content of TNF-α was evaluated via ELISA assay. RESULTS Tolerance to antinociceptive effect of morphine was developed after 7 days of morphine treatment. The concurrent administration of glucosamine (500, 1000 and 2000 mg/kg) with morphine, significantly inhibited tolerance development, on days 7 and 9. In addition, glucosamine ameliorated the naloxone-precipitated opioid withdrawal symptoms (tremor, jumping, teeth chattering, grooming). However, diarrhea was significantly improved only with the dose of 500 mg/kg. Increased mRNA expression of iNOS as well as TNF-α mRNA expression and protein, after both morphine tolerance and withdrawal, were considerably reduced by glucosamine (1000 mg/kg) in the morphine withdrawal animals. CONCLUSION These data support the utility of glucosamine in attenuating both tolerance to nociceptive effects of morphine as well as withdrawal-induced behavioral profile. Anti-oxidant and anti-inflammatory effects are responsible, at least in part, for the protective effects of this drug.
Collapse
Affiliation(s)
- Faezeh Basiri
- Student Research Committee, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Abolfazl Rad
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Davood Mahdian
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Mehdi Molavi
- Departement of Internal Medicine, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Bahareh Amin
- Cellular and Molecular Research Center, Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran.
| |
Collapse
|
21
|
Lin AH, Liu MH, Ko HK, Perng DW, Lee TS, Kou YR. Menthol Cigarette Smoke Induces More Severe Lung Inflammation Than Non-menthol Cigarette Smoke Does in Mice With Subchronic Exposure - Role of TRPM8. Front Physiol 2018; 9:1817. [PMID: 30618827 PMCID: PMC6305452 DOI: 10.3389/fphys.2018.01817] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 12/05/2018] [Indexed: 01/27/2023] Open
Abstract
In smokers with chronic obstructive pulmonary disease, more severe lung inflammation is associated with menthol cigarette smoking compared to non-menthol cigarette smoking. However, the mechanisms remain unclear. Menthol is an activator of transient receptor potential melastatin-8 (TRPM8), which is also sensitive to reactive oxygen species (ROS). Our recent in vitro study demonstrated that the extracts of menthol cigarette smoke (M-CS) can induce greater ROS-sensitive, TRPM8-mediated, mitogen-activated protein kinase (MAPK)-dependent inflammatory responses in lung epithelial cells than the extracts of non-menthol cigarette smoke (Non-M-CS) can. In this study, we tested the hypothesis that M-CS can induce more severe lung inflammation than Non-M-CS can via the additional action of menthol in M-CS on epithelial and lung TRPM8 in mice. Compared with Non-M-CS exposure, subchronic M-CS exposure for 7 days up-regulated the epithelial and lung expression of TRPM8, induced more vigorous activation of epithelial and lung MAPKs, and caused more severe lung inflammation. The MAPK activation was evidenced by the increased expression of phosphor-extracellular signal-regulated and phosphor-c-Jun N-terminal kinases. The lung inflammation was evidenced by pathohistological findings and increases in several inflammatory indices. Moreover, treatment with a TRPM8 antagonist (N-(3-aminopropyl)-2-{[(3-methylphenyl)methyl]oxy}-N-(2-thienylmethyl)benzamide; AMTB) greatly suppressed the MAPK activation and lung inflammation induced by Non-M-CS and M-CS, and the residual responses to these two types of CS did not differ. Conversely, the levels of biomarkers of acute CS exposure (20 min), including carboxyhemoglobin and cotinine (a nicotine metabolite) in blood plasma, and superoxide and hydrogen peroxide (two major types of ROS) in bronchoalveolar lavage fluid, did not show significant differences in the mice with Non-M-CS and M-CS exposure. We concluded that M-CS could induce greater TRPM8-mediated activation of MAPKs and lung inflammation than Non-M-CS could in mice with subchronic exposure. The augmented inflammatory effects of M-CS are unlikely due to a larger total amount of CS inhaled, but may be caused by an additional stimulation of epithelial and lung TRPM8 by menthol in M-CS. A common stimulant (presumably ROS) generated by both CS types may also stimulate TRPM8, activate MAPKs, and induce lung inflammation because treatment with AMTB could reduce these responses to Non-M-CS.
Collapse
Affiliation(s)
- An-Hsuan Lin
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Meng-Han Liu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsin-Kuo Ko
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Diahn-Warng Perng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzong-Shyuan Lee
- Graduate Institute and Department of Physiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu Ru Kou
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
22
|
Hsu HT, Tseng YT, Wong WJ, Liu CM, Lo YC. Resveratrol prevents nanoparticles-induced inflammation and oxidative stress via downregulation of PKC-α and NADPH oxidase in lung epithelial A549 cells. Altern Ther Health Med 2018; 18:211. [PMID: 29986680 PMCID: PMC6038342 DOI: 10.1186/s12906-018-2278-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/02/2018] [Indexed: 11/25/2022]
Abstract
Background Exposure to carbon black nanoparticles (CBNPs), a well-known industrial production, promotes pulmonary toxicity through inflammation and oxidative stress. Recent studies show that some polyphenols exert their antioxidant properties through regulation of protein kinase C-α (PKC-α) and NADPH oxidase (Nox) signaling. Resveratrol, a dietary polyphenol in fruits, possesses various health beneficial effects including anti-inflammatory and antioxidative properties. In this study, we aimed to elucidate the involvement of PKC-α and Nox in CBNPs-induced inflammation and oxidative stress, and to investigate the protective effects of resveratrol on CBNP-induced inflammation and oxidative stress in human lung epithelial A549 cells. Methods The production of reactive oxygen species (ROS) and the change of mitochondrial membrane potential (ΔΨm) were measured by flow cytometry. Nitric oxide (NO) was measured using the Griess reagent, and prostaglandin E2 (PGE2) production was detected by ELISA, while protein expressions were measured by Western blotting analysis. Results In lung epithelial A549 cells, CBNPs significantly enhanced oxidative stress by upregulation of Nox2 and membrane expression of p67phox accompanied with increase of ROS production. CBNPs also increased inflammatory factors, including iNOS, COX-2, NO and PGE2. However, resveratrol attenuated the above effects induced by CBNPs in A549 cells; additionally, CBNPs-induced activation of PKC-α was observed. We found that PKC-α inhibitor (Gö6976) could attenuate CBNPs-induced inflammation by down-regulation of ROS, NO and PGE2 production in A549 cells, suggesting PKC-α might be involved in CBNPs-induced oxidative stress and inflammation. Our results also found resveratrol was able to inhibit protein expression of PKC-α induced by CBNPs. Moreover, ROS scavenger (NAC) and Nox inhibitor (DPI) attenuated CBNPs-induced expressions of iNOS and COX-2. DPI could also attenuate CBNPs-induced ROS, NO and PGE2 production. Conclusions Resveratrol attenuated CBNPs-induced oxidative and inflammatory factors in lung epithelial A549 cells, at least in part via inhibiting PKC-α- and Nox-related signaling.
Collapse
|
23
|
Castellani S, Trapani A, Spagnoletta A, di Toma L, Magrone T, Di Gioia S, Mandracchia D, Trapani G, Jirillo E, Conese M. Nanoparticle delivery of grape seed-derived proanthocyanidins to airway epithelial cells dampens oxidative stress and inflammation. J Transl Med 2018; 16:140. [PMID: 29792199 PMCID: PMC5966913 DOI: 10.1186/s12967-018-1509-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/08/2018] [Indexed: 12/05/2022] Open
Abstract
Background Chronic respiratory diseases, whose one of the hallmarks is oxidative stress, are still incurable and need novel therapeutic tools and pharmaceutical agents. The phenolic compounds contained in grape are endowed with well-recognized anti-oxidant, anti-inflammatory, anti-cancer, and anti-aging activities. Considering that natural anti-oxidants, such as proanthocyanidins, have poor water solubility and oral bioavailability, we have developed a drug delivery system based on solid lipid nanoparticles (SLN), apt to encapsulate grape seed extract (GSE), containing proanthocyanidins. Methods Plain, 6-coumarin (6-Coum), DiR- and GSE-loaded SLN were produced with the melt-emulsion method. Physicochemical characterization of all prepared SLN was determined by photon correlation spectroscopy and laser Doppler anemometry. MTT assay (spectrophotometry) and propidium iodide (PI) assay (cytofluorimetry) were used to assess cell viability. Flow cytometry coupled with cell imaging was performed for assessing apoptosis and necrosis by Annexin V/7-AAD staining (plain SLE), cell internalization (6-Coum-SLN) and reactive oxygen species (ROS) production (SLN-GSE). NF-κB nuclear translocation was studied by immunofluorescence. In vivo bio-imaging was used to assess lung deposition and persistence of aerosolized DiR-loaded SLN. Results Plain SLN were not cytotoxic when incubated with H441 airway epithelial cells, as judged by both PI and MTT assays as well as by apoptosis/necrosis evaluation. 6-Coum-loaded SLN were taken up by H441 cells in a dose-dependent fashion and persisted into cells at detectable levels up to 16 days. SLN were detected in mice lungs up to 6 days. SLN-GSE possessed 243 nm as mean diameter, were negatively charged, and stable in size at 37 °C in Simulated Lung Fluid up to 48 h and at 4 °C in double distilled water up to 2 months. The content of SLN in proanthocyanidins remained unvaried up to 2 months. GSE-loaded SLN determined a significant reduction in ROS production when added 24–72 h before the stimulation with hydrogen peroxide. Interestingly, while at 24 h free GSE determined a higher decrease of ROS production than SLN-GSE, the contrary was seen at 48 and 72 h. Similar results were observed for NF-κB nuclear translocation. Conclusions SLN are a biocompatible drug delivery system for natural anti-oxidants obtained from grape seed in a model of oxidative stress in airway epithelial cells. They feature stability and long-term persistence inside cells where they release proanthocyanidins. These results could pave the way to novel anti-oxidant and anti-inflammatory therapies for chronic respiratory diseases. Electronic supplementary material The online version of this article (10.1186/s12967-018-1509-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- S Castellani
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - A Trapani
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - A Spagnoletta
- Laboratory "BioProducts and BioProcesses", ENEA Research Centre Trisaia, Rotondella, MT, Italy
| | - L di Toma
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - T Magrone
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari "Aldo Moro", Bari, Italy
| | - S Di Gioia
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - D Mandracchia
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - G Trapani
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - E Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari "Aldo Moro", Bari, Italy
| | - M Conese
- Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy.
| |
Collapse
|
24
|
McCarty MF, O'Keefe JH, DiNicolantonio JJ. Glucosamine for the Treatment of Osteoarthritis: The Time Has Come for Higher-Dose Trials. J Diet Suppl 2018; 16:179-192. [PMID: 29667462 DOI: 10.1080/19390211.2018.1448920] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Although clinical trials with glucosamine in osteoarthritis have yielded mixed results, leading to doubts about its efficacy, the utility of glucosamine for preventing joint destruction and inflammation is well documented in rodent models of arthritis, including models of spontaneous osteoarthritis. The benefit of oral glucosamine in adjuvant arthritis is markedly dose dependent, likely reflecting a modulation of tissue levels of UDP-N-acetylglucosamine that in turn influences mucopolysaccharide synthesis and the extent of protein O-GlcNAcylation. Importantly, the minimal oral dose of glucosamine that exerts a detectible benefit in adjuvant arthritis achieves plasma glucosamine levels similar to those achieved when the standard clinical dose of glucosamine, 1.5 g daily, is administered as a bolus. The response of plasma glucosamine levels to an increase in glucosamine intake is nearly linear. Remarkably, every published clinical trial with glucosamine has employed the same 1.5 g dose that Rottapharm recommended for its proprietary glucosamine sulfate product decades ago, yet there has never been any published evidence that this dose is optimal with respect to efficacy and side effects. If this dose is on the edge of demonstrable clinical efficacy when experimental design is ideal, then variations in the patient populations targeted, the assessment vehicles employed, and the potency of glucosamine preparations tested could be expected to yield some null results. Failure to employ bolus dosing may also be a factor in the null results observed in the GAIT study and other trials. Clinical studies evaluating the dose dependency of glucosamine's influence on osteoarthritis are long overdue.
Collapse
Affiliation(s)
| | - James H O'Keefe
- b Mid America Heart Institute , University of Missouri-Kansas City , Kansas City , MO , USA
| | | |
Collapse
|
25
|
Hsiao YH, Tseng CM, Su KC, Chen WC, Wu MT, Wu YC, Chang SC, Lee YC, Kou YR, Perng DW. Glycopyrronium bromide inhibits lung inflammation and small airway remodeling induced by subchronic cigarette smoke exposure in mice. Respir Physiol Neurobiol 2018; 249:16-22. [DOI: 10.1016/j.resp.2017.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 02/02/2023]
|
26
|
Campos KKD, Araújo GR, Martins TL, Bandeira ACB, Costa GDP, Talvani A, Garcia CCM, Oliveira LAM, Costa DC, Bezerra FS. The antioxidant and anti-inflammatory properties of lycopene in mice lungs exposed to cigarette smoke. J Nutr Biochem 2017. [DOI: 10.1016/j.jnutbio.2017.06.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
27
|
Shi J, Shan S, Li H, Song G, Li Z. Anti-inflammatory effects of millet bran derived-bound polyphenols in LPS-induced HT-29 cell via ROS/miR-149/Akt/NF-κB signaling pathway. Oncotarget 2017; 8:74582-74594. [PMID: 29088809 PMCID: PMC5650364 DOI: 10.18632/oncotarget.20216] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 06/05/2017] [Indexed: 12/24/2022] Open
Abstract
The pro-inflammatory and anti-inflammatory maladjustment has been acknowledged as one of the chief causations of inflammatory diseases and even cancers. Previous studies showed that plant-derived polyphenolic compounds were the most potent anti-oxidant and anti-inflammatory agents among all natural compounds. The present study indicates that bound polyphenols of inner shell (BPIS) from foxtail millet bran can display anti-inflammatory effects in LPS-induced HT-29 cells and in nude mice. Mechanistically, BPIS restrained the level of various pro-inflammatory cytokines (IL-1β, IL-6, IL-8), and enhanced the expression level of anti-inflammatory cytokine (IL-10) by blocking the nuclear factor-kappaB (NF-κB)-p65 nuclear translocation. Further, we found the elevated miR-149 expression by BPIS-induced ROS accumulation, directly targeted the Akt expression to block NF-κB nuclear translocation. Taken together, these novel findings provide new insights into the development of BPIS as an anti-inflammatory agent via the signaling cascade of ROS/miR-149/Akt/NF-κB axis.
Collapse
Affiliation(s)
- Jiangying Shi
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, PR China
| | - Shuhua Shan
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, PR China
| | - Hanqing Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, PR China.,College of Life Science, Shanxi University, Taiyuan 030006, China
| | - Guisheng Song
- Department of Medicine, Division of Gastroenterology, University of Minnesota Medical School, Minneapolis, Minnesota, MN 55455, USA
| | - Zhuoyu Li
- Institute of Biotechnology, Key Laboratory of Chemical Biology and Molecular Engineering of National Ministry of Education, Shanxi University, Taiyuan 030006, PR China.,College of Life Science, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
28
|
Yang HL, Korivi M, Chen CH, Peng WJ, Chen CS, Li ML, Hsu LS, Liao JW, Hseu YC. Antrodia camphorata attenuates cigarette smoke-induced ROS production, DNA damage, apoptosis, and inflammation in vascular smooth muscle cells, and atherosclerosis in ApoE-deficient mice. ENVIRONMENTAL TOXICOLOGY 2017; 32:2070-2084. [PMID: 28370894 DOI: 10.1002/tox.22422] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 03/08/2017] [Accepted: 03/19/2017] [Indexed: 06/07/2023]
Abstract
Cigarette smoke exposure activates several cellular mechanisms predisposing to atherosclerosis, including oxidative stress, dyslipidemia, and vascular inflammation. Antrodia camphorata, a renowned medicinal mushroom in Taiwan, has been investigated for its antioxidant, anti-inflammatory, and antiatherosclerotic properties in cigarette smoke extracts (CSE)-treated vascular smooth muscle cells (SMCs), and ApoE-deficient mice. Fermented culture broth of Antrodia camphorata (AC, 200-800 µg/mL) possesses effective antioxidant activity against CSE-induced ROS production. Treatment of SMCs (A7r5) with AC (30-120 µg/mL) remarkably ameliorated CSE-induced morphological aberrations and cell death. Suppressed ROS levels by AC corroborate with substantial inhibition of CSE-induced DNA damage in AC-treated A7r5 cells. We found CSE-induced apoptosis through increased Bax/Bcl-2 ratio, was substantially inhibited by AC in A7r5 cells. Notably, upregulated SOD and catalase expressions in AC-treated A7r5 cells perhaps contributed to eradicate the CSE-induced ROS generation, and prevents DNA damage and apoptosis. Besides, AC suppressed AP-1 activity by inhibiting the c-Fos/c-Jun expressions, and NF-κB activation through inhibition of I-κBα degradation against CSE-stimulation. This anti-inflammatory property of AC was accompanied by suppressed CSE-induced VEGF, PDGF, and EGR-1 overexpressions in A7r5 cells. Furthermore, AC protects lung fibroblast (MRC-5) cells from CSE-induced cell death. In vivo data showed that AC oral administration (0.6 mg/d/8-wk) prevents CSE-accelerated atherosclerosis in ApoE-deficient mice. This antiatherosclerotic property was associated with increased serum total antioxidant status, and decreased total cholesterol and triacylglycerol levels. Thus, Antrodia camphorata may be useful for prevention of CSE-induced oxidative stress and diseases. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 2070-2084, 2017.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Mallikarjuna Korivi
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Cheng-Hsien Chen
- Department of Applied Chemistry, Chao Yang University of Technology, Taichung, Taiwan
| | - Wei-Jung Peng
- Department of Applied Chemistry, Chao Yang University of Technology, Taichung, Taiwan
| | - Chee-Shan Chen
- Department of Applied Chemistry, Chao Yang University of Technology, Taichung, Taiwan
| | - Mei-Ling Li
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Li-Sung Hsu
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung, Taiwan
| | - You-Cheng Hseu
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
29
|
Rac1 signaling regulates cigarette smoke-induced inflammation in the lung via the Erk1/2 MAPK and STAT3 pathways. Biochim Biophys Acta Mol Basis Dis 2017; 1863:1778-1788. [DOI: 10.1016/j.bbadis.2017.04.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 03/24/2017] [Accepted: 04/16/2017] [Indexed: 02/06/2023]
|
30
|
Zhuang L, Chen LF, Zhang YB, Liu Z, Xiao XH, Tang W, Wang GC, Song WJ, Li YL, Li MM. Watsonianone A from Rhodomyrtus tomentosa Fruit Attenuates Respiratory-Syncytial-Virus-Induced Inflammation In Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:3481-3489. [PMID: 28436225 DOI: 10.1021/acs.jafc.7b00537] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Respiratory syncytial virus (RSV) is one of the most common respiratory pathogens. Immoderate inflammation plays a great role in causing RSV-induced diseases. In the present study, watsonianone A, isolated from the fruit of Rhodomyrtus tomentosa (Ait.) Hassk, was found to show a good inhibitory effect on RSV-induced NO production, with a half-maximal inhibitory concentration of 37.2 ± 1.6 μM. Enzyme-linked immunosorbent assay and fluorescence quantitative polymerase chain reaction analyses indicated that watsonianone A markedly reduced both mRNA and protein levels of tumor necrosis factor α, interleukin 6, and monocyte chemoattractant protein 1 in RSV-infected RAW264.7 cells. Mechanistically, watsonianone A inhibited nuclear factor κB (NF-κB) activation by suppressing IκBα phosphorylation. Further analysis revealed that watsonianone A activated the thioredoxin system and decreased intracellular reactive oxygen species (ROS) levels, which are closely associated with NF-κB activation in RSV-infected cells. These results reveal that watsonianone A can attenuate RSV-induced inflammation via the suppression of ROS-sensitive inflammatory signaling.
Collapse
Affiliation(s)
- Ling Zhuang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Li-Feng Chen
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Yu-Bo Zhang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Zhong Liu
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Xu-Hui Xiao
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Wei Tang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Guo-Cai Wang
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Wen-Jun Song
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Yao-Lan Li
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| | - Man-Mei Li
- Institute of Traditional Chinese Medicine and Natural Products, College of Pharmacy, and ‡Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University , Guangzhou, Guangdong 510632, People's Republic of China
| |
Collapse
|
31
|
Lin AH, Liu MH, Ko HKB, Perng DW, Lee TS, Kou YR. Inflammatory Effects of Menthol vs. Non-menthol Cigarette Smoke Extract on Human Lung Epithelial Cells: A Double-Hit on TRPM8 by Reactive Oxygen Species and Menthol. Front Physiol 2017; 8:263. [PMID: 28496415 PMCID: PMC5406458 DOI: 10.3389/fphys.2017.00263] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 04/10/2017] [Indexed: 11/21/2022] Open
Abstract
Clinical studies suggest that smokers with chronic obstructive pulmonary disease who use menthol cigarettes may display more severe lung inflammation than those who smoke non-menthol cigarette. However, the mechanisms for this difference remain unclear. Menthol is a ligand of transient receptor potential melastatin-8 (TRPM8), a Ca2+-permeant channel sensitive to reactive oxygen species (ROS). We previously reported that exposure of human bronchial epithelial cells (HBECs) to non-menthol cigarette smoke extract (Non-M-CSE) triggers a cascade of inflammatory signaling leading to IL-8 induction. In this study, we used this in vitro model to compare the inflammatory effects of menthol cigarette smoke extract (M-CSE) and Non-M-CSE and delineate the mechanisms underlying the differences in their impacts. Compared with Non-M-CSE, M-CSE initially increased a similar level of extracellular ROS, suggesting the equivalent oxidant potency. However, M-CSE subsequently produced more remarkable elevations in intracellular Ca2+, activation of the mitogen-activated protein kinases (MAPKs)/nuclear factor-κB (NF-κB) signaling, and IL-8 induction. The extracellular ROS responses to both CSE types were totally inhibited by N-acetyl-cysteine (NAC; a ROS scavenger). The intracellular Ca2+ responses to both CSE types were also totally prevented by NAC, AMTB (a TRPM8 antagonist), or EGTA (an extracellular Ca2+ chelator). The activation of the MAPK/NF-κB signaling and induction of IL-8 to both CSE types were suppressed to similar levels by NAC, AMTB, or EGTA. These results suggest that, in addition to ROS generated by both CSE types, the menthol in M-CSE may act as another stimulus to further activate TRPM8 and induce the observed responses. We also found that menthol combined with Non-M-CSE induced greater responses of intracellular Ca2+ and IL-8 compared with Non-M-CSE alone. Moreover, we confirmed the essential role of TRPM8 in these responses to Non-M-CSE or M-CSE and the difference in these responses between the both CSE types using HBECs with TRPM8 knockdown and TRPM8 knockout, and using HEK293 cells transfected with hTRPM8. Thus, compared with exposure to Non-M-CSE, exposure to M-CSE induced greater TRPM8-mediated inflammatory responses in HBECs. These augmented effects may be due to a double-hit on lung epithelial TRPM8 by ROS generated from CSE and the menthol in M-CSE.
Collapse
Affiliation(s)
- An-Hsuan Lin
- Department of Physiology, School of Medicine, National Yang-Ming UniversityTaipei, Taiwan
| | - Meng-Han Liu
- Department of Physiology, School of Medicine, National Yang-Ming UniversityTaipei, Taiwan
| | - Hsin-Kuo B Ko
- Department of Chest Medicine, Taipei Veterans General HospitalTaipei, Taiwan
| | - Diahn-Warng Perng
- Department of Chest Medicine, Taipei Veterans General HospitalTaipei, Taiwan
| | - Tzong-Shyuan Lee
- Department of Physiology, School of Medicine, National Yang-Ming UniversityTaipei, Taiwan
| | - Yu Ru Kou
- Department of Physiology, School of Medicine, National Yang-Ming UniversityTaipei, Taiwan
| |
Collapse
|
32
|
Lee JW, Park JW, Kwon OK, Lee HJ, Jeong HG, Kim JH, Oh SR, Ahn KS. NPS2143 Inhibits MUC5AC and Proinflammatory Mediators in Cigarette Smoke Extract (CSE)-Stimulated Human Airway Epithelial Cells. Inflammation 2017; 40:184-194. [PMID: 27866297 DOI: 10.1007/s10753-016-0468-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mucus overproduction is a fundamental hallmark of COPD that is caused by exposure to cigarette smoke. MUC5AC is one of the main mucin genes expressed in the respiratory epithelium, and its transcriptional upregulation often correlates with increased mucus secretion. Calcium-sensing receptor (CaSR) antagonists have been reported to possess anti-inflammatory effects. The purpose of the present study was to investigate the protective role of NPS2143, a selective CaSR antagonist on cigarette smoke extract (CSE)-stimulated NCI-H292 mucoepidermoid human lung cells. Treatment of NPS2143 significantly inhibited the expression of MUC5AC in CSE-stimulated H292 cells. NPS2143 reduced the expression of MMP-9 in CSE-stimulated H292 cells. NPS2143 also decreased the release of proinflammatory cytokines such as IL-6 and TNF-α in CSE-stimulated H292 cells. Furthermore, NPS2143 attenuated the activation of MAPKs (JNK, p38, and ERK) and inhibited the nuclear translocation of NF-κB in CSE-stimulated H292 cells. These results indicate that NPS2143 had a therapeutic potential in COPD.
Collapse
Affiliation(s)
- Jae-Won Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea
| | - Ji-Won Park
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea
| | - Ok-Kyoung Kwon
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, 305-764, Republic of Korea
| | - Hee Jae Lee
- Department of Pharmacology, College of Medicine, Kangwon National University, Chuncheon, Kangwon, 200-701, Republic of Korea
| | - Hye Gwang Jeong
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon, 305-764, Republic of Korea
| | - Jae-Hong Kim
- Department of Life Sciences and Biotechnology, Korea University, 5-1 Anam-dong, Sungbuk-gu, Seoul, 136-701, Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea.
| | - Kyoung-Seop Ahn
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Chungju-si, Chungbuk, 363-883, Republic of Korea.
| |
Collapse
|
33
|
Nie Y, Huang C, Zhong S, Wortley MA, Luo Y, Luo W, Xie Y, Lai K, Zhong N. Cigarette smoke extract (CSE) induces transient receptor potential ankyrin 1(TRPA1) expression via activation of HIF1αin A549 cells. Free Radic Biol Med 2016; 99:498-507. [PMID: 27480844 DOI: 10.1016/j.freeradbiomed.2016.07.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 07/28/2016] [Accepted: 07/28/2016] [Indexed: 11/26/2022]
Abstract
We previously found that transient receptor potential ankyrin 1 (TRPA1) in guinea pig tracheal epithelial cells was elevated after 14 days of cigarette smoke (CS) exposure. However, the mechanism underlying CS-induced TRPA1 expression remains unknown. Here, we explored whether cigarette smoke extract (CSE)-induced TRPA1 expression is related with modulation of HIF1α in A549 cells. Our results showed that CSE increased TRPA1 expression in A549 cells, decreased Iκ B, PHD2, and HDAC2, and increased ROS release and nuclear translocation of NF-κ B and HIF1α. Moreover, HIF1α siRNA and/or MG132 (a proteasome inhibitor) pretreatment significantly inhibited CSE-induced TRPA1 expression and HIF1α nuclear translocation in A549 cells. However, HIF1α siRNA pretreatment did not affect CSE-induced NF-κ B nuclear translocation, suggesting that CSE-induced TRPA1 expression in A549 cells is directly mediated by HIF1α, but not by NF-κ B. Similar to CSE treatment, treatment of A549 cells with LPS caused significant increases in nuclear translocation of NF-κ B and HIF1α mRNA expression, but did not alter TRPA1 mRNA expression. However, pretreatment with PHD2 siRNA did result in increased TRPA1 mRNA expression in LPS-treated A549 cells; an effect that was inhibited by SN50 (a NF-κ B inhibitor). It suggests a role for NF-κ B to indirectly regulate TRPA1 mRNA expression via modulating HIF1α mRNA transcription. In addition, treatment cells with HDAC2 siRNA plus 2%CSE resulted in increased HIF1α nuclear translocation and TRPA1 expression, which was significantly inhibited by MG132 and HIF1α siRNA. These results suggest that HDAC2 indirectly modulates TRPA1 expression by promoting the DNA-binding activity of HIF1α. These findings show that CSE increases TRPA1 expression in airway epithelial cells by directly activating HIF1α, and that this increase in TRPA1 expression is indirectly regulated via NF-κ B, PHD2 and HDAC2 modulation of HIF1α activity.
Collapse
Affiliation(s)
- Yichu Nie
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No 151 YanJiang Road, Yuexiu Dist., Guangzhou 510120, People's Republic of China; Respiratory Pharmacology Group, Airway Disease Section, National Heart & Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom
| | - Chuqin Huang
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No 151 YanJiang Road, Yuexiu Dist., Guangzhou 510120, People's Republic of China
| | - Shan Zhong
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No 151 YanJiang Road, Yuexiu Dist., Guangzhou 510120, People's Republic of China
| | - Michael A Wortley
- Respiratory Pharmacology Group, Airway Disease Section, National Heart & Lung Institute, Imperial College London, London SW7 2AZ, United Kingdom
| | - Yulong Luo
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No 151 YanJiang Road, Yuexiu Dist., Guangzhou 510120, People's Republic of China
| | - Wei Luo
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No 151 YanJiang Road, Yuexiu Dist., Guangzhou 510120, People's Republic of China
| | - Yanqing Xie
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No 151 YanJiang Road, Yuexiu Dist., Guangzhou 510120, People's Republic of China
| | - Kefang Lai
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No 151 YanJiang Road, Yuexiu Dist., Guangzhou 510120, People's Republic of China.
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Diseases, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, No 151 YanJiang Road, Yuexiu Dist., Guangzhou 510120, People's Republic of China
| |
Collapse
|
34
|
Molecular mechanisms and biomedical applications of glucosamine as a potential multifunctional therapeutic agent. Life Sci 2016; 152:21-9. [PMID: 27012765 DOI: 10.1016/j.lfs.2016.03.028] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 03/10/2016] [Accepted: 03/11/2016] [Indexed: 12/30/2022]
Abstract
Glucosamine and its acetylated derivative, N-acetyl glucosamine, are naturally occurring amino sugars found in human body. They are important components of glycoproteins, proteoglycans and glycosaminoglycans. Scientific studies have supported that glucosamine has the beneficial pharmacological effects to relieve osteoarthritis symptoms. Glucosamine can also be as a promising candidate for the prevention and/or treatment of some other diseases due to its anti-oxidant and anti-inflammatory activities. Most of its function is exerted by modulation of inflammatory responses especially through Nuclear Factor-κB (NF-κB) that can control inflammatory cytokine production and cell survival. In this review, we present a concise update on additional new therapeutic applications of glucosamine including treatment of cardiovascular disease, neurological deficits, skin disorders, cancer and the molecular mechanistic rationale for these uses. This article will also examine safety profile and adverse effects of glucosamine in human.
Collapse
|
35
|
Caro CA, Lillo L, Valenzuela FJ, Cabello G, Lang E, Vallejos D, Castillo C. Oxidation of melatonin on a glassy carbon electrode modified with metallic glucosamines. Synthesis and characterization. J Solid State Electrochem 2016. [DOI: 10.1007/s10008-015-3066-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
36
|
Yang J, Yu HM, Zhou XD, Huang HP, Han Z, Kolosov VP, Perelman JM. Cigarette smoke induces mucin hypersecretion and inflammatory response through the p66shc adaptor protein-mediated mechanism in human bronchial epithelial cells. Mol Immunol 2016; 69:86-98. [PMID: 26608927 DOI: 10.1016/j.molimm.2015.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/02/2015] [Accepted: 11/03/2015] [Indexed: 10/22/2022]
Abstract
The p66Shc adaptor protein is a newly recognized mediator of mitochondrial dysfunction and might play a role in cigarette smoke (CS)-induced airway epithelial cell injury. CS can induce an excessive amount of reactive oxygen species (ROS) generation, which can cause mitochondrial depolarization and injury through the oxidative stress-mediated Serine36 phosphorylation of p66Shc. The excessive production of ROS can trigger an inflammatory response and mucin hypersecretion by enhancing the transcriptional activity of pro-inflammatory cytokines and mucin genes. Therefore, we speculate that p66Shc plays an essential role in airway epithelial cell injury and the process of mucin generation in CS-induced chronic inflammatory airway diseases. Our present study focuses on the role of p66Shc in ROS generation, and on the resulting mitochondrial dysfunction, inflammatory response and mucus hypersecretion in CS-stimulated human bronchial epithelial cells (16HBE). We found that CS disturbed the mitochondrial function by increasing the level of phosphorylated p66Shc in these cells and that the effects were significantly reduced by silencing p66Shc. Conversely, the ectopic overexpression of wild-type p66Shc enhanced these effects. We also found that high levels of ROS inhibited FOXO3a transcriptional activity, which led to NF-κB activation. Subsequently, activated NF-κB promoted pro-inflammatory cytokine production and mucin hypersecretion. Thus, manipulating p66Shc might offer a new therapeutic modality with which to treat chronic inflammatory airway diseases.
Collapse
Affiliation(s)
- J Yang
- Division of Respiratory Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - H M Yu
- Division of Geriatrics Medicine, First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - X D Zhou
- Division of Respiratory Medicine, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China; Division of Respiratory Medicine, Affiliated Hospital of Hainan Medical University, Haikou, China.
| | - H P Huang
- Division of Respiratory Medicine, Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zh Han
- Division of Respiratory Medicine, Affiliated Hospital of Hainan Medical University, Haikou, China
| | - V P Kolosov
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Siberian Branch, Russian Academy of Medical Sciences, Russian Federation
| | - J M Perelman
- Far Eastern Scientific Center of Physiology and Pathology of Respiration, Siberian Branch, Russian Academy of Medical Sciences, Russian Federation
| |
Collapse
|
37
|
Lung Epithelial TRPA1 Transduces the Extracellular ROS into Transcriptional Regulation of Lung Inflammation Induced by Cigarette Smoke: The Role of Influxed Ca²⁺. Mediators Inflamm 2015; 2015:148367. [PMID: 26504357 PMCID: PMC4609484 DOI: 10.1155/2015/148367] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 04/02/2015] [Indexed: 12/27/2022] Open
Abstract
The mechanism underlying the inflammatory role of TRPA1 in lung epithelial cells (LECs) remains unclear. Here, we show that cigarette smoke extract (CSE) sequentially induced several events in LECs. The Ca2+ influx was prevented by decreasing extracellular reactive oxygen species (ROS) with the scavenger N-acetyl-cysteine, removing extracellular Ca2+ with the chelator EGTA, or treating with the TRPA1 antagonist HC030031. NADPH oxidase activation was abolished by its inhibitor apocynin, EGTA, or HC030031. The increased intracellular ROS was halted by apocynin, N-acetyl-cysteine, or HC030031. The activation of the MAPKs/NF-κB signaling was suppressed by EGTA, N-acetyl-cysteine, or HC030031. IL-8 induction was inhibited by HC030031 or TRPA1 siRNA. Additionally, chronic cigarette smoke (CS) exposure in wild-type mice induced TRPA1 expression in LECs and lung tissues. In CS-exposure trpa1−/− mice, the increased BALF level of ROS was similar to that of CS-exposure wild-type mice; yet lung inflammation was lessened. Thus, in LECs, CSE may initially increase extracellular ROS, which activate TRPA1 leading to an increase in Ca2+ influx. The increased intracellular Ca2+ contributes to activation of NADPH oxidase, resulting in increased intracellular ROS, which activate the MAPKs/NF-κB signaling leading to IL-8 induction. This mechanism may possibly be at work in mice chronically exposed to CS.
Collapse
|
38
|
Gao W, Li L, Wang Y, Zhang S, Adcock IM, Barnes PJ, Huang M, Yao X. Bronchial epithelial cells: The key effector cells in the pathogenesis of chronic obstructive pulmonary disease? Respirology 2015; 20:722-9. [PMID: 25868842 DOI: 10.1111/resp.12542] [Citation(s) in RCA: 158] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 02/25/2015] [Accepted: 02/27/2015] [Indexed: 01/06/2023]
Abstract
The primary function of the bronchial epithelium is to act as a defensive barrier aiding the maintenance of normal airway function. Bronchial epithelial cells (BEC) form the interface between the external environment and the internal milieu, making it a major target of inhaled insults. However, BEC can also serve as effectors to initiate and orchestrate immune and inflammatory responses by releasing chemokines and cytokines, which recruit and activate inflammatory cells. They also produce excess reactive oxygen species as a result of an oxidant/antioxidant imbalance that contributes to chronic pulmonary inflammation and lung tissue damage. Accumulated mucus from hyperplastic BEC obstructs the lumen of small airways, whereas impaired cell repair, squamous metaplasia and increased extracellular matrix deposition underlying the epithelium is associated with airway remodelling particularly fibrosis and thickening of the airway wall. These alterations in small airway structure lead to airflow limitation, which is critical in the clinical diagnosis of chronic obstructive pulmonary disease (COPD). In this review, we discuss the abnormal function of BEC within a disturbed immune homeostatic environment consisting of ongoing inflammation, oxidative stress and small airway obstruction. We provide an overview of recent insights into the function of the bronchial epithelium in the pathogenesis of COPD and how this may provide novel therapeutic approaches for a number of chronic lung diseases.
Collapse
Affiliation(s)
- Wei Gao
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lingling Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yujie Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Sini Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| | - Peter J Barnes
- Airway Disease Section, National Heart and Lung Institute, Imperial College, London, UK
| | - Mao Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Yao
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
39
|
Ramesh T, Sureka C, Bhuvana S, Begum VH. Oxidative stress in the brain of cigarette smoke-induced noxiousness: neuroprotective role of Sesbania grandiflora. Metab Brain Dis 2015; 30:573-82. [PMID: 25217401 DOI: 10.1007/s11011-014-9614-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/25/2014] [Indexed: 12/20/2022]
Abstract
Cigarette smoking is concerned as a major risk factor in the development of various neurological disorders. Oxidative stress is suggested as a possible contributing factor in the pathogenesis of cigarette smoking-induced toxicity. Therefore, the present study was intended to evaluate the neuroprotective role of Sesbania grandiflora (S. grandiflora) against chronic cigarette smoke induced oxidative damage in rat brain. Adult male Wistar-Kyoto rats were exposed to cigarette smoke for a period of 90 days and consecutively treated with S. grandiflora aqueous suspension (SGAS, 1,000 mg/kg body weight per day by oral gavage) for a period of 3 weeks. Lipid peroxidation and antioxidants status were analyzed in the brain. Rats exposed to cigarette smoke showed significant increase in conjugated diens (CD), hydroperoxides (HP) and malendialdehyde (MDA) levels with concomitant decrease in superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), glutathione-S-transferase (GST) and glucose-6-phosphate dehydrogenase (G6PDH) activities and the levels of reduced glutathione (GSH), vitamin C and vitamin E. Also cigarette smoke-exposure resulted in a marked increase in copper and decrease in zinc, manganese and selenium levels in brain. Administration of SGAS attenuates lipid peroxidation, enhanced the antioxidant status, restored the levels of micronutrients and retained the brain histology. The results of our study indicate that chronic cigarette smoke-exposure accelerates oxidative stress, thereby disquieting the brain defensive mechanism and S. grandiflora protects the brain from the oxidative damage through its biopotency.
Collapse
Affiliation(s)
- Thiyagarajan Ramesh
- Department of Biochemistry, College of Medicine, Salman Bin Abdulaziz University, Al-Kharj, 11942, Kingdom of Saudi Arabia,
| | | | | | | |
Collapse
|
40
|
Hwang JS, Park JW, Nam MS, Cho H, Han IO. Glucosamine enhances body weight gain and reduces insulin response in mice fed chow diet but mitigates obesity, insulin resistance and impaired glucose tolerance in mice high-fat diet. Metabolism 2015; 64:368-79. [PMID: 25516476 DOI: 10.1016/j.metabol.2014.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 10/28/2014] [Accepted: 11/15/2014] [Indexed: 12/14/2022]
Abstract
OBJECTIVE This study investigated the potential of glucosamine (GlcN) to affect body weight gain and insulin sensitivity in mice normal and at risk for developing diabetes. METHODS Male C57BL/6J mice were fed either chow diet (CD) or a high fat diet (HFD) and the half of mice from CD and HFD provided with a solution of 10% (w/v) GlcN. Total cholesterol and nonesterified free fatty acid levels were determined. Glucose tolerance test and insulin tolerance test were performed. HepG2 human hepatoma cells or differentiated 3T3-L1 adipocytes were stimulated with insulin under normal (5 mM) or high glucose (25 mM) conditions. Effect of GlcN on 2-deoxyglucose (2-DG) uptake was determined. JNK and Akt phosphorylation and nucleocytoplasmic protein O-GlcNAcylation were assayed by Western blotting. RESULTS GlcN administration stimulated body weight gain (6.58±0.82 g vs. 11.1±0.42 g), increased white adipose tissue fat mass (percentage of bodyweight, 3.7±0.32 g vs. 5.61±0.34 g), and impaired the insulin response in livers of mice fed CD. However, GlcN treatment in mice fed HFD led to reduction of body weight gain (18.02±0.66 g vs. 16.22±0.96 g) and liver weight (2.27±0.1 vs. 1.85±0.12 g). Furthermore, obesity-induced insulin resistance and impaired Akt insulin signaling in the liver were alleviated by GlcN administration. GlcN inhibited the insulin response under low (5 mM) glucose conditions, whereas it restored the insulin response for Akt phosphorylation under high (25 mM) glucose conditions in HepG2 and 3T3-L1 cells. Uptake of 2-DG increased upon GlcN treatment under 5 mM glucose compared to control, whereas insulin-stimulated 2-DG uptake decreased under 5 mM and increased under 25 mM glucose in differentiated 3T3-L1 cells. CONCLUSION Our results show that GlcN increased body weight gain and reduced the insulin response for glucose maintenance when fed to normal CD mice, whereas it alleviated body weight gain and insulin resistance in HFD mice. Therefore, the current data support the integrative function of the HBP reflecting the nutrient status of lipids or glucose and further implicate the importance of the pathway in insulin signaling for the regulation of metabolism.
Collapse
Affiliation(s)
- Ji-Sun Hwang
- Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Ji-Won Park
- Department of Life Science, Inha University, Incheon, Republic of Korea
| | - Moon-Suk Nam
- Department of Endocrinology & Metabolism, Clinical Pharmacology, College of Medicine, Inha University, Incheon, Republic of Korea
| | - Hyeongjin Cho
- Department of Chemistry, Inha University, Incheon, Republic of Korea
| | - Inn-Oc Han
- Department of Physiology and Biophysics, College of Medicine, Inha University, Incheon, Republic of Korea.
| |
Collapse
|
41
|
Kang N, Chen P, Chen Y, Zeng H, He X, Zhu Y. PRMT6 mediates CSE induced inflammation and apoptosis. Int Immunopharmacol 2015; 24:95-101. [PMID: 25481537 DOI: 10.1016/j.intimp.2014.10.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 10/09/2014] [Accepted: 10/28/2014] [Indexed: 01/19/2023]
Abstract
Cigarette smoke extract (CSE) induces apoptosis and inflammation, but the mechanism is unknown. Arginine methyltransferase (PRMT6) catalyzes the asymmetric di-methylation of histone H3 arginine 2 (H3R2me2a) to control global level transcription. We hypothesized that PRMT6 mediates CSE induced apoptosis and inflammation through H3R2me2a. The apoptosis after CSE treatment in human umbilical vein endothelial cells (HUVECs) was fully measured with real-time reverse transcription PCR, western blotting and Annexin-V staining. Meanwhile, the inflammation in HUVECs after CSE exposure was detected with real-time reverse transcription PCR, western blotting and ELISA. CSE treatment promoted apoptosis and inflammation in HUVECs, coinciding with the decreased protein abundance of PRMT6. Meanwhile, HUVECs transfected with PRMT6 expressing plasmid inhibited the CSE-induced apoptosis and inflammation. Also, the inhibition of PRMT6 promoted the apoptosis and inflammation in HUVECs induced by CSE. Notably, H3R2me2a was associated with the modulation of PRMT6 in CSE induced apoptosis and inflammation in HUVECs. In conclusion, PRMT6 mediates CSE induced apoptosis and inflammation through H3R2me2a in HUVECs.
Collapse
Affiliation(s)
- Naixing Kang
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Ping Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Yan Chen
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China.
| | - Huihui Zeng
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Xue He
- Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan 410011, China
| | - Yingqun Zhu
- Department of Respiratory Medicine, The Third Hospital of Changsha, Changsha, Hunan 410015, China
| |
Collapse
|
42
|
Liu MH, Lin AH, Lu SH, Peng RY, Lee TS, Kou YR. Eicosapentaenoic acid attenuates cigarette smoke-induced lung inflammation by inhibiting ROS-sensitive inflammatory signaling. Front Physiol 2014; 5:440. [PMID: 25452730 PMCID: PMC4231989 DOI: 10.3389/fphys.2014.00440] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/27/2014] [Indexed: 12/16/2022] Open
Abstract
Cigarette smoking causes chronic lung inflammation that is mainly regulated by redox-sensitive pathways. Our previous studies have demonstrated that cigarette smoke (CS) activates reactive oxygen species (ROS)-sensitive mitogen-activated protein kinases (MAPKs)/nuclear factor-κB (NF-κB) signaling resulting in induction of lung inflammation. Eicosapentaenoic acid (EPA), a major type of omega-3 polyunsaturated fatty acid, is present in significant amounts in marine-based fish and fish oil. EPA has been shown to possess antioxidant and anti-inflammatory properties in vitro and in vivo. However, whether EPA has similar beneficial effects against CS-induced lung inflammation remains unclear. Using a murine model, we show that subchronic CS exposure for 4 weeks caused pulmonary inflammatory infiltration (total cell count in bronchoalveolar lavage fluid (BALF), 11.0-fold increase), increased lung vascular permeability (protein level in BALF, 3.1-fold increase), elevated levels of chemokines (11.4–38.2-fold increase) and malondialdehyde (an oxidative stress biomarker; 2.0-fold increase) in the lungs, as well as lung inflammation; all of these CS-induced events were suppressed by daily supplementation with EPA. Using human bronchial epithelial cells, we further show that CS extract (CSE) sequentially activated NADPH oxidase (NADPH oxidase activity, 1.9-fold increase), increased intracellular levels of ROS (3.0-fold increase), activated both MAPKs and NF-κB, and induced interleukin-8 (IL-8; 8.2-fold increase); all these CSE-induced events were inhibited by pretreatment with EPA. Our findings suggest a novel role for EPA in alleviating the oxidative stress and lung inflammation induced by subchronic CS exposure in vivo and in suppressing the CSE-induced IL-8 in vitro via its antioxidant function and by inhibiting MAPKs/NF-κB signaling.
Collapse
Affiliation(s)
- Meng-Han Liu
- Department of Physiology, School of Medicine, National Yang-Ming University Taipei, Taiwan
| | - An-Hsuan Lin
- Department of Physiology, School of Medicine, National Yang-Ming University Taipei, Taiwan
| | - Shing-Hwa Lu
- Department of Urology, Taipei City Hospital, Zhong-Xiao Branch Taipei, Taiwan
| | - Ruo-Yun Peng
- Hsin Sheng Junior College of Medical Care and Management Longtan Township, Taiwan
| | - Tzong-Shyuan Lee
- Department of Physiology, School of Medicine, National Yang-Ming University Taipei, Taiwan
| | - Yu Ru Kou
- Department of Physiology, School of Medicine, National Yang-Ming University Taipei, Taiwan
| |
Collapse
|
43
|
Zhao J, Zhang Z, Dai J, wang L, Zhang C, Ye Y, Li L. Synergistic protective effect of chlorogenic acid, apigenin and caffeic acid against carbon tetrachloride-induced hepatotoxicity in male mice. RSC Adv 2014. [DOI: 10.1039/c4ra07261h] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
44
|
Paeonol attenuates cigarette smoke-induced lung inflammation by inhibiting ROS-sensitive inflammatory signaling. Mediators Inflamm 2014; 2014:651890. [PMID: 25165413 PMCID: PMC4137546 DOI: 10.1155/2014/651890] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 12/20/2022] Open
Abstract
Cigarette smoking causes persistent lung inflammation that is mainly regulated by redox-sensitive pathways. We have previously reported that cigarette smoke (CS) activates reactive oxygen species- (ROS-) sensitive mitogen-activated protein kinases (MAPKs)/nuclear factor-κB (NF-κB) signaling leading to induction of lung inflammation. Paeonol, the main phenolic compound present in the Chinese herb Paeonia suffruticosa, has antioxidant and anti-inflammatory properties. However, whether paeonol has similar beneficial effects against CS-induced lung inflammation remains unclear. Using a murine model, we showed that chronic CS exposure for 4 weeks caused pulmonary inflammatory infiltration, increased lung vascular permeability, elevated lung levels of chemokines, cytokines, and 4-hydroxynonenal (an oxidative stress biomarker), and induced lung inflammation; all of these CS-induced events were suppressed by chronic treatment with paeonol. Using human bronchial epithelial cells (HBECs), we demonstrated that cigarette smoke extract (CSE) sequentially increased extracellular and intracellular levels of ROS, activated the MAPKs/NF-κB signaling, and induced interleukin-8 (IL-8); all these CSE-induced events were inhibited by paeonol pretreatment. Our findings suggest a novel role for paeonol in alleviating the oxidative stress and lung inflammation induced by chronic CS exposure in vivo and in suppressing CSE-induced IL-8 in vitro via its antioxidant function and an inhibition of the MAPKs/NF-κB signaling.
Collapse
|