1
|
Wei H, Zhang Y, Li T, Zhang S, Yin J, Liu Y, Xing L, Bao J, Li J. Intermittent mild cold stimulation alleviates cold stress-induced pulmonary fibrosis by inhibiting the TGF-β1/Smad signaling pathway in broilers. Poult Sci 2024; 103:103246. [PMID: 37980728 PMCID: PMC10685030 DOI: 10.1016/j.psj.2023.103246] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/21/2023] Open
Abstract
To investigate the potential protective effect of intermittent cold stimulation on lung tissues of broilers exposed to acute cold stress (ACS). A total of 384 one-day-old broilers were assigned to 4 experimental groups with 6 replicates of 16 birds each: control (CON) and ACS groups were reared at normal feeding temperature from d 1 to 42; cold treatment groups (CS3+ACS and CS9+ACS) were reared, respectively, at 3°C or 9°C for 5 h on alternate days below the CON group from d 15 to 35. Animals in CS3+ACS, CS9+ACS, and ACS groups were exposed at 10°C for 24 h on d 43. Subsequently, lung tissues were collected to perform histopathological examination and measurement of relevant indexes. The results showed that lung tissues in CS9+ACS and ACS groups exhibited increased inflammatory cell infiltrates and collagen deposition compared to the CON group, while this pathological phenomenon was less pronounced in the CS3+ACS group. Compared to CON group, H2O2 and MDA contents were increased, and the activities of antioxidant enzymes (CAT, SOD, GPx, T-AOC) were reduced in CS9+ACS and ACS group (P < 0.05); mRNA and protein levels of inhibitor of NF-κB, Smad7, matrix metallopeptidase (MMP)-2, MMP9, and antioxidant-related genes were downregulated, whereas mRNA and protein levels of genes related to NF-κB/NLRP3 pathway-regulated inflammation and TGF-β1/Smad pathway-regulated fibrosis were upregulated in cold-stressed broilers (P < 0.05). mRNA levels of heme oxygenase-1, NAD(P)H quinone oxidoreductase-1, and MMP9 were increased in CS3+ACS group (P < 0.05). Moreover, the expression of most antioxidant-related genes was increased, and that of inflammation- and fibrosis-related genes was reduced in CS3+ACS group (P < 0.05). Therefore, cold stress caused oxidative stress and inflammation, leading to pulmonary fibrosis in broilers, whereas intermittent mild cold stimulation at 3°C below normal rearing temperature alleviated fibrosis by inhibiting the TGF-β1/Smad pathway modulated by the Nrf2/HO-1 and NF-κB/NLRP3 signaling pathway. This study suggests that intermittent mild cold stimulation can be a potential strategy to reduce ACS-induced lung damage in broilers.
Collapse
Affiliation(s)
- Haidong Wei
- College of Life Science, Northeast Agricultural University, 150030 Harbin, China
| | - Yong Zhang
- College of Life Science, Northeast Agricultural University, 150030 Harbin, China
| | - Tingting Li
- College of Life Science, Northeast Agricultural University, 150030 Harbin, China
| | - Shijie Zhang
- College of Life Science, Northeast Agricultural University, 150030 Harbin, China
| | - Jingwen Yin
- College of Life Science, Northeast Agricultural University, 150030 Harbin, China
| | - Yuanyuan Liu
- College of Life Science, Northeast Agricultural University, 150030 Harbin, China
| | - Lu Xing
- College of Life Science, Northeast Agricultural University, 150030 Harbin, China
| | - Jun Bao
- College of Animal Science and Technology, Northeast Agricultural University, 150030 Harbin, China; Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, 150030 Harbin, China
| | - Jianhong Li
- College of Life Science, Northeast Agricultural University, 150030 Harbin, China; Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, 150030 Harbin, China.
| |
Collapse
|
2
|
Li J, Cui Z, Wei M, Almutairi MH, Yan P. Omics analysis of the effect of cold normal saline stress through gastric gavage on LPS induced mice. Front Microbiol 2023; 14:1256748. [PMID: 38163070 PMCID: PMC10755949 DOI: 10.3389/fmicb.2023.1256748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/13/2023] [Indexed: 01/03/2024] Open
Abstract
Cold stress is a significant environmental stimulus that negatively affects the health, production, and welfare of animals and birds. However, the specific effects of cold stimulation combined with lipopolysaccharide (LPS) on the mouse intestine remain poorly understood. Therefore, we designed this research to explore the effect of cold stimulation + LPS on mice intestine via microbiome and microbiota sequencing. Forty-eight mice were randomly divided into four experimental groups (n = 12): Control (CC), LPS-induced (CL), cold normal saline-induced (MC) and LPS + cold normal saline-induced (ML). Our results showed body weight was similar among different groups of mice. However, the body weight of mice in groups CC and CL were slightly higher compared to those in groups MC and ML. The results of gene expressions reflected that CL and ML exposure caused gut injury and barrier dysfunction, as evident by decreased ZO-1, OCCLUDIN (P < 0.01), and CASPASE-1 (P < 0.01) expression in the intestine of mice. Moreover, we found that cold stress induced oxidative stress in LPS-challenged mice by increasing malondialdehyde (MDA) accumulation and decreasing the antioxidant capacity [glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), total and antioxidant capacity (T-AOC)]. The cold stress promoted inflammatory response by increased IL-1β in mice treated with cold normal saline + LPS. Whereas, microbiome sequencing revealed differential abundance in four phyla and 24 genera among the mouse groups. Metabolism analysis demonstrated the presence of 4,320 metabolites in mice, with 43 up-regulated and 19 down-regulated in CC vs. MC animals, as well as 1,046 up-regulated and 428 down-regulated in ML vs. CL animals. It is Concluded that cold stress enhances intestinal damage by disrupting the balance of gut microbiota and metabolites, while our findings contribute in improving management practices of livestock in during cold seasons.
Collapse
Affiliation(s)
- Jing Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhihao Cui
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ming Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Mikhlid H. Almutairi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Peishi Yan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
3
|
Ding Z, Chen W, Wu H, Li W, Mao X, Su W, Zhang Y, Lin N. Integrative network fusion-based multi-omics study for biomarker identification and patient classification of rheumatoid arthritis. Chin Med 2023; 18:48. [PMID: 37143094 PMCID: PMC10158004 DOI: 10.1186/s13020-023-00750-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/10/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Cold-dampness Syndrome (RA-Cold) and Hot-dampness Syndrome (RA-Hot) are two distinct groups of rheumatoid arthritis (RA) patients with different clinical symptoms based on traditional Chinese medicine (TCM) theories and clinical empirical knowledge. However, the biological basis of the two syndromes has not been fully elucidated, which may restrict the development of personalized medicine and drug discovery for RA diagnosis and therapy. METHODS An integrative strategy combining clinical transcriptomics, phenomics, and metabolomics data based on clinical cohorts and adjuvant-induced arthritis rat models was performed to identify novel candidate biomarkers and to investigate the biological basis of RA-Cold and RA-Hot. RESULTS The main clinical symptoms of RA-Cold patients are joint swelling, pain, and contracture, which may be associated with the dysregulation of T cell-mediated immunity, osteoblast differentiation, and subsequent disorders of steroid biosynthesis and phenylalanine metabolism. In contrast, the main clinical symptoms of RA-Hot patients are fever, irritability, and vertigo, which may be associated with various signals regulating angiogenesis, adrenocorticotropic hormone release, and NLRP3 inflammasome activation, leading to disorders of steroid biosynthesis, nicotinamide, and sphingolipid metabolism. IL17F, 5-HT, and IL4I1 were identified as candidate biomarkers of RA-Cold, while S1P and GLNS were identified as candidate biomarkers of RA-Hot. CONCLUSIONS The current study presents the most comprehensive metabonomic and transcriptomic profiling of serum, urine, synovial fluid, and synovial tissue samples obtained from RA-Cold and RA-Hot patients and experimental animal models to date. Through the integration of multi-omics data and clinical independent validation, a list of novel candidate biomarkers of RA-Cold and RA-Hot syndromes were identified, that may be useful in improving RA diagnosis and therapy.
Collapse
Affiliation(s)
- Zihe Ding
- Research Center of Traditional Chinese Medicine Theory and Literatures, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Wenjia Chen
- Research Center of Traditional Chinese Medicine Theory and Literatures, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Hao Wu
- School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Weijie Li
- Research Center of Traditional Chinese Medicine Theory and Literatures, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Xia Mao
- Research Center of Traditional Chinese Medicine Theory and Literatures, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China
| | - Weiwei Su
- School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yanqiong Zhang
- Research Center of Traditional Chinese Medicine Theory and Literatures, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China.
| | - Na Lin
- Research Center of Traditional Chinese Medicine Theory and Literatures, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16, Nanxiaojie, Dongzhimennei, Beijing, 100700, China.
| |
Collapse
|
4
|
Liu J, Wu J, Qiao C, He Y, Xia S, Zheng Y, Lv H. Impact of chronic cold exposure on lung inflammation, pyroptosis and oxidative stress in mice. Int Immunopharmacol 2023; 115:109590. [PMID: 36577159 DOI: 10.1016/j.intimp.2022.109590] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/30/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022]
Abstract
Chronic cold exposure, which is the main inducer of lung diseases in high latitudes, affects production efficiency and restricts the development of aquaculture. Although the relationship between cold exposure and susceptibility to the lungs is widely accepted, but the influence between them has not been fully explored. The aim of this study is to understand the underlying mechanism. In the present study, the mice, which are used to establish cold stress (CS)-induced lung injury model, are exposed to cold temperature (4 °C) for 3 h each day for 4 weeks. The results indicate that the expression of heat shock protein 70 (HSP70) is augmented by cold exposure. In addition, chronic cold exposure aggravate the formation of malondialdehyde (MDA) and lead to a significant decrease in the contents of micrococcus catalase (CAT) and glutathione (GSH). Moreover, chronic cold exposure significantly exacerbates the expression of inflammation- and apoptosis-related proteins. The activation of Bax and caspase-3 are significantly augmented. However, that of Bcl-2 is decreased. These results are different from those in room team. The results show that chronic cold exposure plays an important roles in the activation of multiple signaling pathways, such as pyroptosis-related, inflammation-related and oxidative stress-regulated signaling pathways. In summary, these investigations support that chronic cold exposure increase the risk of lung injury by activating inflammation, oxidative stress and pyroptosis.
Collapse
Affiliation(s)
- Jiahe Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jingjing Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Chunyu Qiao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yuxi He
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Shijie Xia
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yuwei Zheng
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| | - Hongming Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, Heilongjiang Bayi, China; Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural Affairs, Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China.
| |
Collapse
|
5
|
Tran HM, Chen TT, Lu YH, Tsai FJ, Chen KY, Ho SC, Wu CD, Wu SM, Lee YL, Chung KF, Kuo HP, Lee KY, Chuang HC. Climate-mediated air pollution associated with COPD severity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 843:156969. [PMID: 35760178 DOI: 10.1016/j.scitotenv.2022.156969] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/08/2022] [Accepted: 06/21/2022] [Indexed: 06/15/2023]
Abstract
Air pollution has been reported to be associated with chronic obstructive pulmonary disease (COPD). Our study aim was to examine the mediating effects of air pollution on climate-associated health outcomes of COPD patients. A cross-sectional study of 117 COPD patients was conducted in a hospital in Taiwan. We measured the lung function, 6-min walking distance, oxygen desaturation, white blood cell count, and percent emphysema (low attenuation area, LAA) and linked these to 0-1-, 0-3-, and 0-5-year lags of individual-level exposure to relative humidity (RH), temperature, and air pollution. Linear regression models were conducted to examine associations of temperature, RH, and air pollution with severity of health outcomes. A mediation analysis was conducted to examine the mediating effects of air pollution on the associations of RH and temperature with health outcomes. We observed that a 1 % increase in the RH was associated with increases in forced expiratory volume in 1 s (FEV1), eosinophils, and lymphocytes, and a decrease in the total-lobe LAA. A 1 °C increase in temperature was associated with decreases in oxygen desaturation, and right-, left-, and upper-lobe LAA values. Also, a 1 μg/m3 increase in PM2.5 was associated with a decrease in the FEV1 and an increase in oxygen desaturation. A 1 μg/m3 increases in PM10 and PM2.5 was associated with increases in the total-, right-, left, upper-, and lower-lobe (PM2.5 only) LAA. A one part per billion increase in NO2 was associated with a decrease in the FEV1 and an increase in the upper-lobe LAA. Next, we found that NO2 fully mediated the association between RH and FEV1. We found PM2.5 fully mediated associations of temperature with oxygen saturation and total-, right-, left-, and upper-lobe LAA. In conclusion, climate-mediated air pollution increased the risk of decreasing FEV1 and oxygen saturation and increasing emphysema severity among COPD patients. Climate change-related air pollution is an important public health issue, especially with regards to respiratory disease.
Collapse
Affiliation(s)
- Huan Minh Tran
- Ph.D. Program in Global Health and Health Security, College of Public Health, Taipei Medical University, Taipei, Taiwan; Faculty of Public Health, Da Nang University of Medical Technology and Pharmacy, Da Nang, Viet Nam.
| | - Tzu-Tao Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| | - Yueh-Hsun Lu
- Department of Radiology, Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Radiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Feng-Jen Tsai
- Ph.D. Program in Global Health and Health Security, College of Public Health, Taipei Medical University, Taipei, Taiwan.
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Shu-Chuan Ho
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Chih-Da Wu
- Department of Geomatics, National Cheng Kung University, Tainan, Taiwan; National Institute of Environmental Health Sciences, National Health Research Institutes, Miaoli, Taiwan.
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| | - Yueh-Lun Lee
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, UK.
| | - Han-Pin Kuo
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| | - Hsiao-Chi Chuang
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan; School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
6
|
Zhang D, Ma S, Wang L, Ma H, Wang W, Xia J, Liu D. Min pig skeletal muscle response to cold stress. PLoS One 2022; 17:e0274184. [PMID: 36155652 PMCID: PMC9512212 DOI: 10.1371/journal.pone.0274184] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/18/2022] [Indexed: 11/18/2022] Open
Abstract
The increased sensitivity of pigs to ambient temperature is due to today's intensive farming. Frequent climate disasters increase the pressure on healthy pig farming. Min pigs are an indigenous pig breed in China with desirable cold resistance characteristics, and hence are ideal for obtaining cold-resistant pig breeds. Therefore, it is important to discover the molecular mechanisms that are activated in response to cold stress in the Min pig. Here, we conducted a transcriptomic analysis of the skeletal muscle of Min pigs under chronic low-temperature acclimation (group A) and acute short cold stress (group B). Cold exposure caused more genes to be upregulated. Totals of 125 and 96 differentially expressed genes (DEGs) were generated from groups A and B. Sixteen common upregulated DEGs were screened; these were concentrated in oxidative stress (SRXN1, MAFF), immune and inflammatory responses (ITPKC, AREG, MMP25, FOSL1), the nervous system (RETREG1, GADD45A, RCAN1), lipid metabolism (LRP11, LIPG, ITGA5, AMPD2), solute transport (SLC19A2, SLC28A1, SLCO4A1), and fertility (HBEGF). There were 102 and 73 genes that were specifically differentially expressed in groups A and B, respectively. The altered mRNAs were enriched in immune, endocrine, and cancer pathways. There were 186 and 91 differentially expressed lncRNAs generated from groups A and B. Analysis of the target genes suggested that they may be involved in regulating the MAPK signaling pathway for resistance to cold. The results of this study provide a comprehensive overview of cold exposure-induced transcriptional patterns in skeletal muscle of the Min pig. These results can guide future molecular studies of cold stress response in pigs for improving cold tolerance as a goal in breeding programs.
Collapse
Affiliation(s)
- Dongjie Zhang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, Heilongjiang, People’s Republic of China
| | - Shouzheng Ma
- Department of Animal Science, Northeast Agricultural University, Harbin, Heilongjiang, People’s Republic of China
| | - Liang Wang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, Heilongjiang, People’s Republic of China
| | - Hong Ma
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, Heilongjiang, People’s Republic of China
| | - Wentao Wang
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, Heilongjiang, People’s Republic of China
| | - Jiqao Xia
- Department of Animal Science, Northeast Agricultural University, Harbin, Heilongjiang, People’s Republic of China
| | - Di Liu
- Institute of Animal Husbandry, Heilongjiang Academy of Agricultural Sciences, Harbin, Heilongjiang, People’s Republic of China
- Department of Animal Science, Northeast Agricultural University, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
7
|
Tao J, Hossain MZ, Xu Z, Ho HC, Khan MA, Huang C, Zheng H, Ni J, Fan Y, Bogale D, Su H, Cheng J. Protective effect of pneumococcal conjugate vaccination on the short-term association between low temperatures and childhood pneumonia hospitalizations: Interrupted time-series and case-crossover analyses in Matlab, Bangladesh. ENVIRONMENTAL RESEARCH 2022; 212:113156. [PMID: 35331698 DOI: 10.1016/j.envres.2022.113156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/10/2022] [Accepted: 03/18/2022] [Indexed: 06/14/2023]
Abstract
Studies have shown that ambient extreme temperatures (heat and cold) were associated with an increased risk of childhood pneumonia, but the evidence is very limited in low-middle-income countries. It also remains unknown whether pneumococcal conjugate vaccine (PCV) could prevent temperature-related childhood pneumonia. This study collected data on ambient temperature and hospitalizations for childhood pneumonia in Matlab, Bangladesh from 2012 to 2016. Interrupted time series (ITS) analysis was employed to assess the impact of PCV (10-valent) intervention on childhood pneumonia hospitalizations. A time-stratified case-crossover analysis with a conditional logistic regression was performed to examine the association of childhood pneumonia hospitalizations with extreme temperatures and heatwaves before and after PCV10 intervention. Subgroup analyses were conducted to explore the modification effects of seasons, age, gender, and socioeconomic levels on temperature-related childhood pneumonia hospitalizations. We found that after PCV10 intervention, there was a sharp decrease in hospitalizations for childhood pneumonia (relative risk (RR): 0.59, 95% confidence interval (CI): 0.43-0.83). During the study period, heat effects on childhood pneumonia appeared immediately on the current day (odds ratio (OR): 1.28; 95% CI: 1.02-1.60, lag 0), while cold effects appeared 4 weeks later (OR: 1.53, 95% CI: 1.06-2.22, lag 28). Importantly, cold effects decreased significantly after PCV10 (p-value<0.05), but heat and heatwave effects increased after PCV10 (p-value<0.05). Particularly, children from families with a middle or low socioeconomic level, boys, and infants were more susceptible to heat-related pneumonia. This study suggests that PCV10 intervention in Bangladesh may help decrease cold-related not heat-related childhood pneumonia.
Collapse
Affiliation(s)
- Junwen Tao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Major Autoimmune Disease, Hefei, China
| | - Mohammad Zahid Hossain
- International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Zhiwei Xu
- School of Public Health, Faculty of Medicine, University of Queensland, 288 Herston Road, Herston, QLD, 4006, Australia
| | - Hung Chak Ho
- Department of Urban Planning and Design, The University of Hong Kong, Hong Kong, China
| | - Md Alfazal Khan
- Matlab Health Research Centre, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Cunrui Huang
- Vanke School of Public Health, Tsinghua University, Beijing, China
| | - Hao Zheng
- Department of Environmental Health, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Major Autoimmune Disease, Hefei, China
| | - Yinguan Fan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Major Autoimmune Disease, Hefei, China
| | - Daniel Bogale
- College of Health Sciences, Arsi University, Asela, Ethiopia
| | - Hong Su
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Major Autoimmune Disease, Hefei, China
| | - Jian Cheng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Major Autoimmune Disease, Hefei, China.
| |
Collapse
|
8
|
Liu C, Peng Q, Wei L, Li Z, Zhang X, Wu Y, Wang J, Zheng X, Wen Y, Zheng R, Yan Q, Ye Q, Ma J. Deficiency of Lactoferrin aggravates lipopolysaccharide-induced acute inflammation via recruitment macrophage in mice. Biometals 2022; 36:549-562. [PMID: 35650365 PMCID: PMC9159647 DOI: 10.1007/s10534-022-00398-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/14/2022] [Indexed: 11/26/2022]
Abstract
Lactoferrin (Lf), a multiple functional natural immune protein, is widely distributed in mammalian milk and glandular secretions (bile, saliva, tears and nasal mucosal secretions, etc.). In the previous study, we found that Lf plays an anti-inflammatory and anti-tumorigenesis role in AOM/DSS (azoxymethane/dextran sulfate sodium) induced mouse colitis-associated colon cancer model.
Although we found that Lf has anti-inflammatory effects in chronic inflammation, its specific role and mechanisms in acute inflammation have not been clarified. Here, we reported that the expression levels of Lf were significantly increased when the organism was infected by Gram-negative bacteria. We then explored the role and potential mechanism of Lf in lipopolysaccharide (LPS)-induced acute inflammation. In the LPS-induced acute abdominal inflammation model, Lf deficiency aggravated inflammatory response and promoted macrophage chemotaxis to the inflammation site. Lf inhibited macrophage chemotaxis by suppressing the expression of macrophage-associated chemokines Ccl2 and Ccl5. Highly activated NF-κB signaling in Lf−/− mice was responsible for the high expression of Ccl2 and Ccl5. Our results suggested that the anti-inflammatory effect of Lf offers a new potential treatment for acute inflammatory diseases.
Collapse
Affiliation(s)
- Can Liu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Qiu Peng
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Lingyu Wei
- Department of Pathology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Zhengshuo Li
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Xiaoyue Zhang
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Yangge Wu
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Jia Wang
- Department of Immunology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Xiang Zheng
- Department of Pathology, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Yuqing Wen
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Run Zheng
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Qun Yan
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Qiurong Ye
- Department of Pathology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China.
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China.
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China.
| |
Collapse
|
9
|
CHENG Q, MAO Y, DING X. Establishment of a mouse pneumonia model under cold stress. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.52721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
10
|
Zhu L, Wu H, Ma Z, Dong D, Yang Z, Tian J. Astaxanthin ameliorates lipopolysaccharide-induced acute lung injury via inhibition of inflammatory reactions and modulation of the SOCS3/JAK2/STAT3 signaling pathways in mice. Food Funct 2022; 13:11638-11651. [DOI: 10.1039/d2fo02182j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The results showed that astaxanthin had a protective effect on LPS-induced acute lung injury in mice, and its protective mechanism was through activating the SOCS3/JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Li Zhu
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Huihui Wu
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Zhenbo Ma
- Medical Imaging Center, Taian City Central Hospital, No. 29, Longtan Road, Taian 271000, China
| | - Decheng Dong
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Ze Yang
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| | - Jing Tian
- The Department of Blood Transfusion, The Second Affiliated Hospital of Shandong First Medical University, No. 366. Taishan Road, Taian 271000, China
| |
Collapse
|
11
|
Veremchuk LV, Mineeva EE, Vitkina TI, Grigorieva EA, Gvozdenko TA, Golokhvast KS. The response ranges of pulmonary function and the impact criteria of weather and industrial influence on patients with asthma living in Vladivostok. JOURNAL OF ENVIRONMENTAL HEALTH SCIENCE & ENGINEERING 2020; 18:235-242. [PMID: 32399235 PMCID: PMC7203380 DOI: 10.1007/s40201-020-00458-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 02/10/2020] [Indexed: 06/11/2023]
Abstract
PURPOSE To determine the response of pulmonary function (PF) to the influence of environmental factors in patients with different levels of asthma control. MATERIALS AND METHODS Patients with controlled (136 people) and uncontrolled (96 people) asthma living in the conditions of monsoon climate and technogenic pollution in Vladivostok were examined. Discriminant analysis that provides the basis for dividing initial data into classes, as according to standards and expert estimates, was used to calculate ranges of PF response in asthma patients. The selection of discriminant functions with the highest values of constant and coefficient made it possible to identify the optimal quantitative ranges. RESULTS Analysis of the discriminant value of Wilks' lambda (α) has shown that the intensity of PF response to climatic and technogenic factors varies depending on level of disease control (controlled asthma - α = 0.67-0.79, uncontrolled asthma - α =0.05-0.44). The criteria and ranges of PF response also differ depending on level of disease control. In controlled asthma, PF response reflects an adaptive-compensatory dependence. The reaction to the environmental factors is rather weak; therefore, it could be detected by only more sensitive examination method (body plethysmography). In uncontrolled asthma, the response to the influence of environment quality is active and could be clearly identified by spirography. CONCLUSIONS The climatic and technogenic environment of Vladivostok causes strong pathogenic impact on patients with uncontrolled asthma. The effects of dust fraction 0-1 μm, deeply penetrating into respiratory organs, and day-to-day variability of wind speed, which induces weather sensitivity, are particularly adverse.
Collapse
Affiliation(s)
- Lyudmila V. Veremchuk
- Vladivostok Branch of Federal State Budgetary Science Institution, Far Eastern Scientific Center of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russian Federation
| | - Elena E. Mineeva
- Vladivostok Branch of Federal State Budgetary Science Institution, Far Eastern Scientific Center of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russian Federation
| | - Tatyana I. Vitkina
- Vladivostok Branch of Federal State Budgetary Science Institution, Far Eastern Scientific Center of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russian Federation
| | - Elena A. Grigorieva
- Institute for Complex Analysis of Regional Problems Far Eastern Branch of Russian Academy of Sciences, Birobidzhan, Russian Federation
| | - Tatyana A. Gvozdenko
- Vladivostok Branch of Federal State Budgetary Science Institution, Far Eastern Scientific Center of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russian Federation
| | - Kirill S. Golokhvast
- Vladivostok Branch of Federal State Budgetary Science Institution, Far Eastern Scientific Center of Physiology and Pathology of Respiration, Institute of Medical Climatology and Rehabilitative Treatment, Vladivostok, Russian Federation
- Far Eastern Federal University, Vladivostok, Russian Federation
| |
Collapse
|
12
|
Yu Z, Liu X, Chen H, Zhu L. Naringenin-Loaded Dipalmitoylphosphatidylcholine Phytosome Dry Powders for Inhaled Treatment of Acute Lung Injury. J Aerosol Med Pulm Drug Deliv 2020; 33:194-204. [PMID: 32176552 DOI: 10.1089/jamp.2019.1569] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background: Acute lung injury is a severe respiratory disorder characterized by overwhelming lung inflammation. Dipalmitoylphosphatidylcholine (DPPC) is the major lipid component of pulmonary surfactant, which here acts as a carrier delivery system for drugs, while also preserving surface tension in the lung. The clinical development of naringenin (NG) is limited by its low solubility and bioavailability. Methods: Novel NG-loaded DPPC phytosomes for dry powder inhalation (NPDPIs) were prepared by solvent evaporation and a freeze-drying method. The particle size, electric potential, in vitro release, and lung deposition were characterized. A rat model of acute lung injury was established and used for pharmacodynamic evaluations. Results: A mixture of NG/DPPC 1:2 (w/w) formed stable phytosomes with the addition of appropriate ethanol. The phytosomes had high complexation efficiency (92.1% ± 1.87%) with NG, a small mean size (150.8 ± 6.9 nm), and a high zeta potential (20.97 ± 0.55 mV). NPDPIs composed of mannitol/DPPC/NG (4:2:1, w/w/w) presented a satisfactory appearance, good fluidity, quick reconstitution to naringenin phytosomes (NGPs), and small (167.2 nm) reconstituted NGPs. The aerodynamic diameter (12.48 μm) and fine particle fraction (23.90%) were suitable for pulmonary delivery by inhalation. The in vivo NPDPIs demonstrated efficacy in a rat model of acute lung injury. NPDPIs significantly inhibited the phosphorylation of P38 in the mitogen-activated protein kinase pathway and suppressed oxidative stress. Surprisingly, the DPPC vehicle exhibited potential effects against acute lung injury by protecting respiratory function. Conclusions: NPDPIs were developed for sustained drug release, promoting pulmonary bioavailability of drug and protecting against acid-induced acute lung injury in rats by pulmonary delivery. NPDPIs are a promising dry powder inhaler for clinical application in acute lung injury.
Collapse
Affiliation(s)
- Zicheng Yu
- Institute of Clinical Pharmacy and Pharmacology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyan Liu
- Department of Pharmacy, Shanghai United Family Pudong Hospital, Shanghai, China
| | - Hongjun Chen
- Institute of Clinical Pharmacy and Pharmacology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lifei Zhu
- Institute of Clinical Pharmacy and Pharmacology, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Amentoflavone ameliorates cold stress-induced inflammation in lung by suppression of C3/BCR/NF-κB pathways. BMC Immunol 2019; 20:49. [PMID: 31888465 PMCID: PMC6937961 DOI: 10.1186/s12865-019-0331-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/22/2019] [Indexed: 12/16/2022] Open
Abstract
Background Cold stress, which may lead to local and systemic injury, is reported to be related to the immune system, especially the complement system. At present, the lack of effective treatment is a critical issue. Amentoflavone (AF), which can inhibit cold stress-induced inflammation in lung by multiple mechanisms, is the main therapeutic ingredient in plants of the genus Selaginella. Results In the current study, we found that cold could induce lung inflammation related to the complement system and its downstream pathways. AF treatment significantly inhibited lung inflammation from cold exposure. We presented evidence that AF can bind to complement component 3 (C3) to regulate inflammation-related pathways involving Lck/Yes novel tyrosine kinase (Lyn), protein kinase B (Akt), nuclear factor-κB (NF-κB) and immune factors. Moreover, 30 mg/kg of AF caused significantly greater improvement than 15 mg/kg in reducing the level of C3 in lung tissue. Conclusions AF can protect lung tissue from cold exposure. The protective effect may be achieved by inhibition of C3 and negative regulation of the B cell receptor (BCR)/NF-κB signaling pathways and high mobility group box 1 (HMGB1), which ultimately ameliorates the inflammatory response.
Collapse
|
14
|
Chen H, Li M. [Role of mammalian target of rapamycin activation in menthol-induced expressions of airway inflammation-related factors in human bronchial epithelial cells in vitro]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:1344-1349. [PMID: 31852644 DOI: 10.12122/j.issn.1673-4254.2019.11.12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the role of mammalian target of rapamycin (mTOR) activation in menthol-induced expression of airway inflammation- related factors in human bronchial epithelial cells and explore its mechanism. METHODS Cultured human bronchial epithelial cells (BEAS-2B) were divided into normal control group, menthol group, rapamycin group, and menthol+rapamycin group with corresponding treatments. The cell viability was measured with CCK-8 method. The mRNA levels of transient receptor potential melastatin 8 (TRPM8), tumor necrosis factor (TNF)-α and interleukin (IL)-1β were detected by RT-PCR, and the protein expressions of phosphorylated mTOR (p-mTOR), TRPM8, TNF-α and IL-1β were determined using Western blotting. The intracellular Ca2+ fluorescence intensity was measured by flow cytometry. RESULTS Compared with the normal control cells, menthol- treated cells showed significantly increased TNF-α, IL-1β, and p-mTOR expression and elevated intracellular Ca2+ concentration (P < 0.05), and the rapamycin-treated cells exhibited significantly decreased p-mTOR expression (P < 0.05). No significant difference was found in TNF-α, IL-1β or intracellular Ca2+ concentration between the normal control and rapamycin-treated cells (P>0.05). Compared with the menthol-treated cells, the cells treated with both menthol and rapamycin showed significantly decreased TNF- α, IL-1β, and p-mTOR expression and obviously lowered intracellular Ca2+ concentration (P < 0.05). CONCLUSIONS Menthol promotes the expressions of airway inflammationrelated factors IL-1β and TNF-α possibly by activating mTOR to cause the increase of intracellular Ca2+ concentration.
Collapse
Affiliation(s)
- Haibo Chen
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Minchao Li
- Department of Respiratory Medicine, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
15
|
Luo B, Shi H, Zhang K, Wei Q, Niu J, Wang J, Hammond SK, Liu S. Cold stress provokes lung injury in rats co-exposed to fine particulate matter and lipopolysaccharide. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 168:9-16. [PMID: 30384172 DOI: 10.1016/j.ecoenv.2018.10.064] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/11/2018] [Accepted: 10/16/2018] [Indexed: 06/08/2023]
Abstract
Cold exposure aggravates respiratory diseases, which are also influenced by the exposures to particulate matter and endotoxin in the air. The aim of this study was to investigate the potential interactions among cold stress, fine particulate matter (PM2.5, particles with aerodynamic diameter of 2.5 µm or less) and lipopolysaccharide (LPS, pure chemical form of endotoxin) on rat lung and to explore the related possible mechanisms of the interactions. Wistar rats were randomly grouped to be exposed to, 1) normal saline (0.9% NaCl), 2) PM2.5, 3) LPS, and 4) PM2.5 and LPS (PM2.5 + LPS) through intratracheal instillation under cold stress (0 °C) and normal temperature (20 °C). Lung function, lung tissue histology, inflammatory response and oxidative stress levels were measured to examine the lung injury and to investigate the potential mechanisms. Exposure to PM2.5 or LPS substantially changed pulmonary function [indicated by peak inspiratory flow (PIF) and peak expiratory flow (PEF)], inflammatory cytokine levels [indicated by interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α)] and lung histology, compared to the non-exposed groups. Exposure to PM2.5 + LPS under cold stress induced the most significant changes, including the increases of IL-6, TNF-α and thiobarbituric acid-reactive substances (TBARS), the decreases of PIF and PEF and more severe lung injury, among all exposure scenarios. Glutathione peroxidase activity and, nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) were found to be suppressed under cold stress, whereas Nrf2 and HO-1 levels were observed to be upregulated by exposure to PM2.5 or LPS under normal temperature. In conclusion, cold stress may aggravate the lung injury in rats induced by simultaneous exposure to PM2.5 and LPS. The progress may involve the suppressing of Nrf2/HO-1 signal pathway.
Collapse
Affiliation(s)
- Bin Luo
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou 730000, China; Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley 94720, USA.
| | - Hongxia Shi
- Health Management Center, Lanzhou University the Second Hospital, Lanzhou 730030, China
| | - Kai Zhang
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Qiaozhen Wei
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Jingping Niu
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Junling Wang
- Institute of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Sally Katharine Hammond
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley 94720, USA
| | - Sa Liu
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley 94720, USA; Environmental & Occupational Health Sciences, School of Health Sciences, Purdue University, West Lafayette 47907, USA.
| |
Collapse
|
16
|
He R, Li Y, Zhou L, Su X, Li Y, Pan P, Hu C. miR-146b overexpression ameliorates lipopolysaccharide-induced acute lung injury in vivo and in vitro. J Cell Biochem 2018; 120:2929-2939. [PMID: 30500983 DOI: 10.1002/jcb.26846] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 03/09/2018] [Indexed: 12/15/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is a type of acute lung injury (ALI), which causes high morbidity and mortality. So far, effective clinical treatment of ARDS is still limited. Recently, miR-146b has been reported to play a key role in inflammation. In the present study, we evaluated the functional role of miR-146b in ARDS using the murine model of lipopolysaccharide (LPS)-induced ALI. The miR-146b expression could be induced by LPS stimulation, and miR-146b overexpression was required in the maintenance of body weight and survival of ALI mice; after miR-146b overexpression, LPS-induced lung injury, pulmonary inflammation, total cell and neutrophil counts, proinflammatory cytokines, and chemokines in bronchial alveolar lavage (BAL) fluid were significantly reduced. The promotive effect of LPS on lung permeability through increasing total protein, albumin and IgM in BAL fluid could be partially reversed by miR-146b overexpression. Moreover, in murine alveolar macrophages, miR-146b overexpression reduced LPS-induced TNF-α and interleukin (IL)-1β releasing. Taken together, we demonstrated that miR-146b overexpression could effectively improve the LPS-induced ALI; miR-146b is a promising target in ARDS treatment.
Collapse
Affiliation(s)
- Ruoxi He
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Li Zhou
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoli Su
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanyuan Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Pinhua Pan
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Zhang K, Guo L, Wei Q, Song Q, Liu J, Niu J, Zhang L, Ruan Y, Luo B. COPD rat model is more susceptible to cold stress and PM 2.5 exposure and the underlying mechanism. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 241:26-34. [PMID: 29793105 DOI: 10.1016/j.envpol.2018.05.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 05/11/2018] [Accepted: 05/11/2018] [Indexed: 05/25/2023]
Abstract
The purpose of this study is to verify the hypothesis that chronic obstructive pulmonary disease (COPD) model rat is more susceptible to cold stress and fine particulate matter (PM2.5) exposure than the healthy rat, and explore the related mechanism. COPD rat model, established with cigarette smoke and lipopolysaccharide intratracheal instillation, were exposed to cold stress (0 °C) and PM2.5 (0, 3.2, 12.8 mg/ml). After that, the levels of superoxide dismutase, inducible nitric oxide synthase (iNOS), tumor necrosis factor alpha (TNF-α), monocyte chemotactic protein 1 (MCP-1) and angiotensin Ⅱ (Ang-Ⅱ) in lung were measured, as well as the expression levels of lung 8-hydroxy-2-deoxyguanosine (8-OHdG), nuclear factor kappa B (NF-κB), heme-oxygenase-1 (HO-1) and nuclear factor erythroid-2-related factor 2 (Nrf2). There were significant positive relationships between PM2.5 and lung level of iNOS, TNF-α, MCP-1 and Ang-Ⅱ, lung function and pathologic damage in COPD rats. The HO-1, NF-κB and 8-OHdG were found highly expressed in COPD rat lung, particularly at the higher PM2.5 dose of cold stress groups, while Nrf2 was found declined. Thus, COPD rats may be more susceptible to cold stress and PM2.5 exposure. Cold stress may aggravate PM2.5-induced toxic effects in the lung of COPD rats through increasing Ang-Ⅱ/NF-κB signaling pathway and suppressing Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Kai Zhang
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Lei Guo
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Qiaozhen Wei
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Quanquan Song
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Jiangtao Liu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Jingping Niu
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Li Zhang
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Ye Ruan
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China
| | - Bin Luo
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou, Gansu 730000, People's Republic of China.
| |
Collapse
|
18
|
Wu G, Xu G, Chen DW, Gao WX, Xiong JQ, Shen HY, Gao YQ. Hypoxia Exacerbates Inflammatory Acute Lung Injury via the Toll-Like Receptor 4 Signaling Pathway. Front Immunol 2018; 9:1667. [PMID: 30083155 PMCID: PMC6064949 DOI: 10.3389/fimmu.2018.01667] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 07/04/2018] [Indexed: 12/13/2022] Open
Abstract
Acute lung injury (ALI) is characterized by non-cardiogenic diffuse alveolar damage and often leads to a lethal consequence, particularly when hypoxia coexists. The treatment of ALI remains a challenge: pulmonary inflammation and hypoxia both contribute to its onset and progression and no effective prevention approach is available. Here, we aimed to investigate the underlying mechanism of hypoxia interaction with inflammation in ALI and to evaluate hypoxia-inducible factor 1 alpha (HIF-1α)—the crucial modulator in hypoxia—as a potential therapeutic target against ALI. First, we developed a novel ALI rat model induced by a combined low-dose of lipopolysaccharides (LPS) with acute hypoxia. Second, we used gene microarray analysis to evaluate the inflammatory profiles of bronchi alveolar lavage fluid cells of ALI rats. Third, we employed an alveolar macrophage cell line, NR8383 as an in vitro system together with a toll-like receptor 4 (TLR4) antagonist TAK-242, to verify our in vivo findings from ALI animals. Finally, we tested the therapeutic effects of HIF-1α augmentation against inflammation and hypoxia in ALI. We demonstrated that (i) LPS upregulated inflammatory genes, tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6), in the alveolar macrophages of ALI rats, which were further enhanced when ALI combined with hypoxia; (ii) hypoxia exposure could further enhance the upregulation of alveolar macrophageal TLR4 that was noticed in LPS-induced inflammatory ALI, conversely, TLR4 antagonist TAK-242 could suppress the macrophageal expression of TLR4 and inflammatory cytokines, including TNF-α, IL-1β, and IL-6, suggesting that the TLR4 signaling pathway as a central link between inflammation and hypoxia in ALI; (iii) manipulation of HIF-1α in vitro could suppress TLR4 expression induced by combined LPS and hypoxia, via suppressing promoter activity of the TLR4 gene; (iv) preconditioning augmentation of HIF-1α in vivo by HIF hydroxylase inhibitor, DMOG excreted protection against inflammatory, and hypoxic processes in ALI. Together, we see that hypoxia can exacerbate inflammation in ALI via the activation of the TLR4 signaling pathway in alveolar macrophages and predispose impairment of the alveolar-capillary barrier in the development of ALI. Targeting HIF-1α can suppress TLR4 expression and macrophageal inflammation, suggesting the potential therapeutic and preventative value of HIF-1α/TLR4 crosstalk pathway in ALI.
Collapse
Affiliation(s)
- Gang Wu
- College of High Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical University, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Gang Xu
- College of High Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical University, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - De-Wei Chen
- Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China.,Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing, China
| | - Wen-Xiang Gao
- College of High Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical University, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| | - Jian-Qiong Xiong
- Intensive Care Unit, Southwest Hospital, Army Medical University, Chongqing, China
| | - Hai-Ying Shen
- Robert Stone Dow Laboratories, Legacy Research Institute, Legacy Health, Portland, OR, United States
| | - Yu-Qi Gao
- College of High Altitude Military Medicine, Institute of Medicine and Hygienic Equipment for High Altitude Region, Army Medical University, Chongqing, China.,Key Laboratory of High Altitude Medicine, People's Liberation Army, Chongqing, China
| |
Collapse
|
19
|
Moon SC, Joo SY, Chung TW, Choi HJ, Park MJ, Choi HJ, Bae SJ, Kim KJ, Kim CH, Joo M, Ha KT. Abiotic stress of ambient cold temperature regulates the host receptivity to pathogens by cell surfaced sialic acids. Biochem Biophys Res Commun 2016; 476:159-66. [PMID: 27181350 DOI: 10.1016/j.bbrc.2016.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/04/2016] [Indexed: 12/19/2022]
Abstract
Ambient cold temperature, as an abiotic stress, regulates the survival, stability, transmission, and infection of pathogens. However, the effect of cold temperature on the host receptivity to the pathogens has not been fully studied. In this study, the expression of terminal α-2,3- and α-2,6-sialic acids were increased in murine lung tissues, especially bronchial epithelium, by exposure to cold condition. The expression of several sialyltransferases were also increased by exposure to cold temperature. Furthermore, in human bronchial epithelial BEAS-2B cells, the expressions of α-2,3- and α-2,6-sialic acids, and mRNA levels of sialyltransferases were increased in the low temperature condition at 33 °C. On the other hand, the treatment of Lith-Gly, a sialyltransferase inhibitor, blocked the cold-induced expression of sialic acids on surface of BEAS-2B cells. The binding of influenza H1N1 hemagglutinin (HA) toward BEAS-2B cells cultured at low temperature condition was increased, compared to 37 °C. In contrast, the cold-increased HA binding was blocked by treatment of lithocholicglycine and sialyl-N-acetyl-D-lactosamines harboring α-2,3- and α-2,6-sialyl motive. These results suggest that the host receptivity to virus at cold temperature results from the expressions of α-2,3- and α-2,6-sialic acids through the regulation of sialyltransferase expression.
Collapse
Affiliation(s)
- Seong-Cheol Moon
- School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Su-Yeon Joo
- School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Tae-Wook Chung
- School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hee-Jung Choi
- School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Mi-Ju Park
- School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Hee-Jin Choi
- School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Sung-Jin Bae
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Keuk-Jun Kim
- Department of Clinical Pathology, TaeKyeung University, Gyeongsan, 38547, Republic of Korea
| | - Cheorl-Ho Kim
- Department of Biological Science, Sungkyunkwan University, Suwon, Kyunggi-do 16419, Republic of Korea
| | - Myungsoo Joo
- School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Ki-Tae Ha
- School of Korean Medicine and Korean Medicine Research Center for Healthy Aging, Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|