1
|
Zhou JX, Qin XD, Liu X, He WT, Zeeshan M, Dharmage SC, Perret J, Bui D, Zhang YT, Sun MK, Huang JW, Liang LX, Dong GH, Zhou Y. Exposure-effect of PFOS and PFOA on lung function: An integrated approach with epidemiological, cellular, and animal studies. ENVIRONMENTAL RESEARCH 2025; 272:121175. [PMID: 39983955 DOI: 10.1016/j.envres.2025.121175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Perfluorooctanesulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) are increasingly recognized for their adverse impact on human health, particularly on lung function. However, current research results are inconsistent and molecular mechanisms remain unclear, with no studies combining epidemiological, in vivo and in vitro investigations. Our population-based study revealed that PFOS and PFOA exposure is negatively associated with lung function. In vitro, PFOS and PFOA exposure significantly downregulated SP-B mRNA and protein levels, and SP-B expression was restored by overexpression of HSD17B1. PFOS induced hypermethylation and downregulated expression of HSD17B1 in tandem with SP-B. Notably, expression of SP-B was restored after treatment with demethyltransferase inhibitor. In vivo studies corroborated these findings, where PFOS exposure resulted in impaired lung function, histopathological changes, and decreased expression of SP-B and HSD17B1 in lung tissues. Our research demonstrates that PFOS downregulates SP-B expression by inducing hypermethylation and downregulating expression of HSD17B1, leading to impaired lung function.
Collapse
Affiliation(s)
- Jia-Xin Zhou
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiao-Di Qin
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xuan Liu
- West China School of PublicHealth and West China FourthHospital, Chengdu, 610041.China
| | - Wan-Ting He
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mohammed Zeeshan
- University of texas health science center at San Antonio, Department of Biochemistry and Structural Biology, 7703 Floyd Curl, San Antonio, TX, 78229, USA
| | - Shyamali C Dharmage
- Centre for Epidemiology and Biostatistics, School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jennifer Perret
- Allergy and Lung Health Unit, Center for Epidemiology and Biostatistics, School of Population & Global Health, The University of Melbourne, Australia Department of Global and Community Health, George Mason University, Melbourne, VIC 3010, Fairfax, VA, 22030, USA
| | - Dinh Bui
- Allergy and Lung Health Unit, Center for Epidemiology and Biostatistics, School of Population & Global Health, The University of Melbourne, Australia Department of Global and Community Health, George Mason University, Melbourne, VIC 3010, Fairfax, VA, 22030, USA
| | - Yun-Ting Zhang
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ming-Kun Sun
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing-Wen Huang
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Xia Liang
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
| | - Guang-Hui Dong
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Yang Zhou
- Joint International Research Laboratory of Environment and Health, Ministry of Education, Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China; State Environmental Protection Key Laboratory of Environmental Pollution Health Risk Assessment, South China Institute of Environmental Sciences, Ministry of Environmental Protection, Guangzhou, 510655, China.
| |
Collapse
|
2
|
Guo Q, Yuan M, Zhang L, Deng M. scPLAN: a hierarchical computational framework for single transcriptomics data annotation, integration and cell-type label refinement. Brief Bioinform 2024; 25:bbae305. [PMID: 38935069 PMCID: PMC11209730 DOI: 10.1093/bib/bbae305] [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: 03/22/2024] [Revised: 05/22/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
MOTIVATION In the past decade, single-cell RNA sequencing (scRNA-seq) has emerged as a pivotal method for transcriptomic profiling in biomedical research. Precise cell-type identification is crucial for subsequent analysis of single-cell data. And the integration and refinement of annotated data are essential for building comprehensive databases. However, prevailing annotation techniques often overlook the hierarchical organization of cell types, resulting in inconsistent annotations. Meanwhile, most existing integration approaches fail to integrate datasets with different annotation depths and none of them can enhance the labels of outdated data with lower annotation resolutions using more intricately annotated datasets or novel biological findings. RESULTS Here, we introduce scPLAN, a hierarchical computational framework designed for scRNA-seq data analysis. scPLAN excels in annotating unlabeled scRNA-seq data using a reference dataset structured along a hierarchical cell-type tree. It identifies potential novel cell types in a systematic, layer-by-layer manner. Additionally, scPLAN effectively integrates annotated scRNA-seq datasets with varying levels of annotation depth, ensuring consistent refinement of cell-type labels across datasets with lower resolutions. Through extensive annotation and novel cell detection experiments, scPLAN has demonstrated its efficacy. Two case studies have been conducted to showcase how scPLAN integrates datasets with diverse cell-type label resolutions and refine their cell-type labels. AVAILABILITY https://github.com/michaelGuo1204/scPLAN.
Collapse
Affiliation(s)
- Qirui Guo
- Center for Quantitative Biology, Peking University, Yiheyuan Road, 100871, Beijing, China
| | - Musu Yuan
- Center for Quantitative Biology, Peking University, Yiheyuan Road, 100871, Beijing, China
| | - Lei Zhang
- Center for Quantitative Biology, Peking University, Yiheyuan Road, 100871, Beijing, China
- Beijing International Center for Mathematical Research, Peking University, Yiheyuan Road, 100871, Beijing, China
- Center for Machine Learning Research, Peking University, Yiheyuan Road, 100871, Beijing, China
| | - Minghua Deng
- Center for Quantitative Biology, Peking University, Yiheyuan Road, 100871, Beijing, China
- School of Mathematical Sciences, Peking University, Yiheyuan Road, 100871, Beijing, China
- Center for Statistical Science, Peking University, Yiheyuan Road, 100871, Beijing, China
| |
Collapse
|
3
|
Jiang YZ, Huang XR, Chang J, Zhou Y, Huang XT. SIRT1: An Intermediator of Key Pathways Regulating Pulmonary Diseases. J Transl Med 2024; 104:102044. [PMID: 38452903 DOI: 10.1016/j.labinv.2024.102044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
Silent information regulator type-1 (SIRT1), a nicotinamide adenine dinucleotide+-dependent deacetylase, is a member of the sirtuins family and has unique protein deacetylase activity. SIRT1 participates in physiological as well as pathophysiological processes by targeting a wide range of protein substrates and signalings. In this review, we described the latest progress of SIRT1 in pulmonary diseases. We have introduced the basic information and summarized the prominent role of SIRT1 in several lung diseases, such as acute lung injury, acute respiratory distress syndrome, chronic obstructive pulmonary disease, lung cancer, and aging-related diseases.
Collapse
Affiliation(s)
- Yi-Zhu Jiang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xin-Ran Huang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jing Chang
- Xiangya Nursing School, Central South University, Changsha, China; Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xiao-Ting Huang
- Xiangya Nursing School, Central South University, Changsha, China.
| |
Collapse
|
4
|
Chen L, Hou Y, Du D, Cui Y, Nie H, Ding Y. MiR-199a-3p in mouse bone marrow mesenchymal stem cell exosomes increases epithelial sodium channel expression in lung injury. Fundam Clin Pharmacol 2022; 36:1011-1019. [PMID: 35667855 DOI: 10.1111/fcp.12807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 05/18/2022] [Accepted: 06/05/2022] [Indexed: 11/28/2022]
Abstract
Acute lung injury (ALI) causes significant morbidity and mortality in critically ill patients, which often presents with extensive accumulation of activated inflammatory cells and diffused alveolar damage accompanied by oxidative stress. Exosomes are nanovesicles, which have notable anti-inflammatory and repair properties, thus alleviating the symptoms of ALI. Epithelial sodium channel (ENaC) is essential for the transepithelial absorption of Na+ and fluid from alveolar spaces. We studied the effects of bone marrow mesenchymal stem cell exosomes (BMSC-exo) on the apoptosis and protein expression of ENaC in primary mouse alveolar epithelial type 2 cells (AT 2 cells). Moreover, the change of miR-199a-3p in AT 2 cells was detected by qRT-PCR, and we studied the regulation of miR-199a-3p on ENaC protein expression. Our results demonstrated that BMSC-exo could not only improve viability and reduce apoptosis in AT 2 cells, but also enhance the expression of ENaC protein and miR-199a-3p. Meanwhile, the upregulation of miR-199a-3p resulted in increased expression of ENaC protein. In summary, the BMSC-exo could participate in the regulation of ENaC through miR-199a-3p originated from BMSC-exo, thereby providing a new pharmacological tool for the treatment of ALI.
Collapse
Affiliation(s)
- Lei Chen
- China Medical University, Shenyang, China
| | - Yapeng Hou
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Danni Du
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Yong Cui
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, China
| | - Hongguang Nie
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yan Ding
- Department of Stem Cells and Regenerative Medicine, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
5
|
Cigarette Smoke Specifically Affects Small Airway Epithelial Cell Populations and Triggers the Expansion of Inflammatory and Squamous Differentiation Associated Basal Cells. Int J Mol Sci 2021; 22:ijms22147646. [PMID: 34299265 PMCID: PMC8305830 DOI: 10.3390/ijms22147646] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/24/2022] Open
Abstract
Smoking is a major risk factor for chronic obstructive pulmonary disease (COPD) and causes remodeling of the small airways. However, the exact smoke-induced effects on the different types of small airway epithelial cells (SAECs) are poorly understood. Here, using air–liquid interface (ALI) cultures, single-cell RNA-sequencing reveals previously unrecognized transcriptional heterogeneity within the small airway epithelium and cell type-specific effects upon acute and chronic cigarette smoke exposure. Smoke triggers detoxification and inflammatory responses and aberrantly activates and alters basal cell differentiation. This results in an increase of inflammatory basal-to-secretory cell intermediates and, particularly after chronic smoke exposure, a massive expansion of a rare inflammatory and squamous metaplasia associated KRT6A+ basal cell state and an altered secretory cell landscape. ALI cultures originating from healthy non-smokers and COPD smokers show similar responses to cigarette smoke exposure, although an increased pro-inflammatory profile is conserved in the latter. Taken together, the in vitro models provide high-resolution insights into the smoke-induced remodeling of the small airways resembling the pathological processes in COPD airways. The data may also help to better understand other lung diseases including COVID-19, as the data reflect the smoke-dependent variable induction of SARS-CoV-2 entry factors across SAEC populations.
Collapse
|
6
|
Ding J, Zhang Z, Huang W, Bi G. Nicotinamide phosphoribosyltransferase inhibitor is a novel therapeutic candidate in LPS-induced neutrophil extracellular traps. Microbiol Immunol 2021; 65:257-264. [PMID: 33871094 DOI: 10.1111/1348-0421.12885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 11/27/2022]
Abstract
Neutrophil extracellular traps (NETs) are beneficial antibacterial defense structures. However, excessive NETs have also been linked to tissue damage and organ dysfunction. LPS and Gram-negative bacteria induce the formation of reactive oxygen species (ROS)-dependent NETs via the JNK pathway. It was found previously that knockdown of nicotinamide phosphoribosyltransferase (NAMPT) upregulates surfactant protein B (SFTPB or SP-B) and attenuates LPS-induced acute lung injury (ALI) via inhibiting JNK activation. This study investigated the effect of FK866, an intracellular NAMPT inhibitor, on the formation of LPS-induced NETs in mouse bronchoalveolar lavage (BAL) neutrophils and in differentiated HL-60 cells. The results show that inhibition of NAMPT by FK866 suppresses NETs formation in BAL neutrophils from the mice exposed to LPS. FK866 also suppresses NETs formation in the differentiated HL-60 cells stimulated with LPS. Additional data indicate that these effects are mediated by suppressing ROS production at least partly via inhibiting JNK activation and depleting NAD(P)H. The utility of inhibition of intracellular NAMPT may be a potential therapy for LPS-induced NETs-related diseases.
Collapse
Affiliation(s)
- Jing Ding
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zuoman Zhang
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weimin Huang
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Guangliang Bi
- Department of Neonatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Novel computational analysis of large transcriptome datasets identifies sets of genes distinguishing chronic obstructive pulmonary disease from healthy lung samples. Sci Rep 2021; 11:10258. [PMID: 33986404 PMCID: PMC8119951 DOI: 10.1038/s41598-021-89762-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/23/2021] [Indexed: 11/08/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) kills over three million people worldwide every year. Despite its high global impact, the knowledge about the underlying molecular mechanisms is still limited. In this study, we aimed to extend the available knowledge by identifying a small set of COPD-associated genes. We analysed different publicly available gene expression datasets containing whole lung tissue (WLT) and airway epithelium (AE) samples from over 400 human subjects for differentially expressed genes (DEGs). We reduced the resulting sets of 436 and 663 DEGs using a novel computational approach that utilises a random depth-first search to identify genes which improve the distinction between COPD patients and controls along the first principle component of the data. Our method identified small sets of 10 and 15 genes in the WLT and AE, respectively. These sets of genes significantly (p < 10–20) distinguish COPD patients from controls with high fidelity. The final sets revealed novel genes like cysteine rich protein 1 (CRIP1) or secretoglobin family 3A member 2 (SCGB3A2) that may underlie fundamental molecular mechanisms of COPD in these tissues.
Collapse
|
8
|
He S, Zhang H, Lu Y, Zhang Z, Zhang X, Zhou N, Hu Z. Nampt promotes osteogenic differentiation and lipopolysaccharide-induced interleukin-6 secretion in osteoblastic MC3T3-E1 cells. Aging (Albany NY) 2021; 13:5150-5163. [PMID: 33535169 PMCID: PMC7950263 DOI: 10.18632/aging.202434] [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: 05/08/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022]
Abstract
The Nicotinamide phosphoribosyltransferase (Nampt)-NAD-Sirt1 pathway modulates processes involved in the pathogenesis of multiple diseases by influencing inflammation. This study aimed to explore the effect of Nampt in osteogenic differentiation and inflammatory response of osteoblastic MC3T3-E1 cells. We developed an in vitro model of lipopolysaccharide (LPS)-induced inflammation and showed that Nampt and Sirt1 were significantly upregulated in LPS-treated MC3T3-E1 cells. LPS induced secretion of the proinflammatory cytokine interleukin-6 (IL-6) and attenuated osteogenic differentiation. Then we transfected cells with adenoviruses to knock down or over express Nampt. Nampt promoted the expression of IL-6, TAK1 and phospho-NF-κB p65 after LPS treatment. Overexpression of Nampt overrode the effect of LPS and rescued LPS-induced inhibition on osteogenic differentiation. FK866, a Nampt inhibitor, had the same inhibitory effect as Nampt knockdown. In addition, Sirt1 suppression by EX527 decreased IL-6 secretion and NF-κB activation without changing the level of Nampt. EX527 also decreased osteogenic differentiation. Incubation with NMN or SRT 1720 also counteract the inhibitory effect of LPS and rescued osteoblast differentiation. Therefore, we demonstrated that Nampt acted both in promoting osteoblast differentiation and in enhancing inflammatory response, mediated by Sirt1 in MC3T3-E1 cells.
Collapse
Affiliation(s)
- Shan He
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.,School of Nursing, Chongqing Medical University, Chongqing 400016, China
| | - Hanxiang Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Yang Lu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhaosi Zhang
- Department of Cerebrovascular Diseases, the First Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Xiang Zhang
- Department of Orthopaedics, Hunan Province People's Hospital, Changsha 410005, China
| | - Nian Zhou
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Zhenming Hu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
9
|
Bossardi Ramos R, Adam AP. Molecular Mechanisms of Vascular Damage During Lung Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:95-107. [PMID: 34019265 DOI: 10.1007/978-3-030-68748-9_6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A variety of pulmonary and systemic insults promote an inflammatory response causing increased vascular permeability, leading to the development of acute lung injury (ALI), a condition necessitating hospitalization and intensive care, or the more severe acute respiratory distress syndrome (ARDS), a disease with a high mortality rate. Further, COVID-19 pandemic-associated ARDS is now a major cause of mortality worldwide. The pathogenesis of ALI is explained by injury to both the vascular endothelium and the alveolar epithelium. The disruption of the lung endothelial and epithelial barriers occurs in response to both systemic and local production of pro-inflammatory cytokines. Studies that evaluate the association of genetic polymorphisms with disease risk did not yield many potential therapeutic targets to treat and revert lung injury. This failure is probably due in part to the phenotypic complexity of ALI/ARDS, and genetic predisposition may be obscured by the multiple environmental and behavioral risk factors. In the last decade, new research has uncovered novel epigenetic mechanisms that control ALI/ARDS pathogenesis, including histone modifications and DNA methylation. Enzyme inhibitors such as DNMTi and HDACi may offer new alternative strategies to prevent or reverse the vascular damage that occurs during lung injury. This review will focus on the latest findings on the molecular mechanisms of vascular damage in ALI/ARDS, the genetic factors that might contribute to the susceptibility for developing this disease, and the epigenetic changes observed in humans, as well as in experimental models of ALI/ADRS.
Collapse
Affiliation(s)
- Ramon Bossardi Ramos
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| | - Alejandro Pablo Adam
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA. .,Department of Ophthalmology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
10
|
Intranasal Application of Budesonide Attenuates Lipopolysaccharide-Induced Acute Lung Injury by Suppressing Nucleotide-Binding Oligomerization Domain-Like Receptor Family, Pyrin Domain-Containing 3 Inflammasome Activation in Mice. J Immunol Res 2019; 2019:7264383. [PMID: 30937316 PMCID: PMC6415278 DOI: 10.1155/2019/7264383] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/20/2018] [Accepted: 11/14/2018] [Indexed: 12/26/2022] Open
Abstract
Aim To investigate the protective effects of budesonide against lipopolysaccharide- (LPS-) induced acute lung injury (ALI) in a murine model and its underlying mechanism. Methods Adult male C57BL/6 mice were divided into three groups: control, ALI, and ALI + budesonide groups. LPS (5 mg/kg) was intratracheally injected to induce ALI in mice. Budesonide (0.5 mg/kg) was intranasally given 1 h before LPS administration in the ALI + budesonide group. Twelve hours after LPS administration, all mice were sacrificed. Hematoxylin-eosin staining and pathological scores were used to evaluate pathological injury. Bronchoalveolar lavage was performed. The numbers of total cells, neutrophils, and macrophages in the bronchoalveolar lavage fluid (BALF) were counted. Enzyme-linked immunosorbent assay was employed to detect the proinflammatory cytokines in BALF and serum, including tumor necrosis factor- (TNF-) α, monocyte chemoattractant protein- (MCP-) 1, and interleukin- (IL-) 1β. The expression of the nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome was detected by western blotting. A lethal dose of LPS (40 mg/kg, intraperitoneally) was injected to evaluate the effects of budesonide on survival rates. Results Budesonide pretreatment dramatically attenuated pathological injury and reduced pathological scores in mice with ALI. Budesonide pretreatment obviously reduced the numbers of total cells, neutrophils, and macrophages in the BALF of mice with ALI. Additionally, budesonide dramatically reduced TNF-α and MCP-1 expression in the BALF and serum of mice with ALI. Budesonide significantly suppressed NLRP3 and pro-caspase-1 expression in the lung and reduced IL-1β content in the BALF, indicating that budesonide inhibited the activation of the NLRP3 inflammasome. Furthermore, we found that budesonide improved the survival rates of mice with ALI receiving a lethal dose of LPS. Conclusion Suppression of NLRP3 inflammasome activation in mice via budesonide attenuated lung injury induced by LPS in mice with ALI.
Collapse
|
11
|
Li X, Islam S, Xiong M, Nsumu NN, Lee MW, Zhang LQ, Ueki Y, Heruth DP, Lei G, Ye SQ. Epigenetic regulation of NfatC1 transcription and osteoclastogenesis by nicotinamide phosphoribosyl transferase in the pathogenesis of arthritis. Cell Death Discov 2019; 5:62. [PMID: 30774990 PMCID: PMC6365567 DOI: 10.1038/s41420-018-0134-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/15/2018] [Accepted: 11/29/2018] [Indexed: 01/17/2023] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) functions in NAD synthesis, apoptosis, and inflammation. Dysregulation of NAMPT has been associated with several inflammatory diseases, including rheumatoid arthritis (RA). The purpose of this study was to investigate NAMPT’s role in arthritis using mouse and cellular models. Collagen-induced arthritis (CIA) in DBA/1J Nampt+/− mice was evaluated by ELISA, micro-CT, and RNA-sequencing (RNA-seq). In vitro Nampt loss-of-function and gain-of-function studies on osteoclastogenesis were examined by TRAP staining, nascent RNA capture, luciferase reporter assays, and ChIP-PCR. Nampt-deficient mice presented with suppressed inflammatory bone destruction and disease progression in a CIA mouse model. Nampt expression was required for the epigenetic regulation of the Nfatc1 promoter and osteoclastogenesis. Finally, RNA-seq identified 690 differentially expressed genes in whole ankle joints which associated (P < 0.05) with Nampt expression and CIA. Selected target was validated by RT-PCR or functional characterization. We have provided evidence that NAMPT functions as a genetic risk factor and a potential therapeutic target to RA.
Collapse
Affiliation(s)
- Xuanan Li
- 1Division of Experimental and Translational Genetics, Children's Mercy, Kansas City, MO 64108 USA.,2Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108 USA.,3Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410005 China
| | - Shamima Islam
- 1Division of Experimental and Translational Genetics, Children's Mercy, Kansas City, MO 64108 USA
| | - Min Xiong
- 1Division of Experimental and Translational Genetics, Children's Mercy, Kansas City, MO 64108 USA
| | - Ndona N Nsumu
- 1Division of Experimental and Translational Genetics, Children's Mercy, Kansas City, MO 64108 USA
| | - Mark W Lee
- 4Department of Chemistry, University of Missouri, Columbia, MO 65211 USA
| | - Li Qin Zhang
- 1Division of Experimental and Translational Genetics, Children's Mercy, Kansas City, MO 64108 USA.,2Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108 USA
| | - Yasuyoshi Ueki
- 5Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO 64108 USA
| | - Daniel P Heruth
- 1Division of Experimental and Translational Genetics, Children's Mercy, Kansas City, MO 64108 USA
| | - Guanghua Lei
- 3Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410005 China
| | - Shui Qing Ye
- 1Division of Experimental and Translational Genetics, Children's Mercy, Kansas City, MO 64108 USA.,2Department of Biomedical and Health Informatics, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108 USA
| |
Collapse
|