1
|
Molinari F, Franco GA, Tranchida N, Di Paola R, Cordaro M. Molecular Mechanism of Action of Endocrine-Disrupting Chemicals on the Respiratory System. Int J Mol Sci 2024; 25:12540. [PMID: 39684250 DOI: 10.3390/ijms252312540] [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: 10/31/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Endocrine-disrupting chemicals (EDCs) are a growing health hazard for humankind and respiratory health in particular. Such chemical compounds are present in the environment and food and may interfere with physiological processes through interference with functions of the endocrine system, making humans more susceptible to various types of diseases. This review aims to discuss the effects of EDCs on the respiratory system. Exposure to EDCs during fetal development and adulthood increases susceptibility to respiratory diseases such as asthma, COPD, and pulmonary fibrosis. EDCs are both multiple and complex in the ways they can act. Indeed, these chemicals may induce oxidative stress, modify cell proliferation and differentiation, interfere with tissue repair, and modulate the inflammatory response. Moreover, EDCs may also break the integrity of the blood-air barrier, allowing noxious substances to penetrate into the lung and thus enhancing the opportunity for infection. In conclusion, the scientific evidence available tends to indicate that EDCs exposure is strongly linked to the initiation of respiratory disease. Further research will be important in discovering the underlying molecular mechanisms and devising preventive and therapeutic measures.
Collapse
Affiliation(s)
- Francesco Molinari
- Department of Veterinary Sciences, University of Messina, Viale SS Annunziata, 98168 Messina, Italy
| | - Gianluca Antonio Franco
- Department of Veterinary Sciences, University of Messina, Viale SS Annunziata, 98168 Messina, Italy
| | - Nicla Tranchida
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale F. Stagno D'Alcontres 31, 98166 Messina, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, Viale SS Annunziata, 98168 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| |
Collapse
|
2
|
Cho IS, Lim J, Chang MS, Lee JH. Impact of the coronavirus disease 2019 pandemic on hospital admissions for idiopathic pulmonary fibrosis: a nationwide population-based study. BMC Pulm Med 2024; 24:430. [PMID: 39217306 PMCID: PMC11365253 DOI: 10.1186/s12890-024-03230-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Previous studies have consistently reported a decrease in hospital admissions for respiratory diseases during the coronavirus disease 2019 (COVID-19) pandemic. However, the impact of the pandemic on idiopathic pulmonary fibrosis (IPF) admissions remains unknown. METHODS This study used data from the Korean National Health Insurance Service database. IPF was defined based on the International Classification of Diseases 10th Revision (ICD-10) and rare intractable disease (RID) codes. The rate of IPF admissions was calculated by dividing the number of IPF admissions by the prevalence of IPF. The rate of IPF admissions during the COVID-19 pandemic (2020-2021) was compared with the mean rate of admissions during the prepandemic period (2017-2019) and presented as the rate ratio (RR). A sensitivity analysis was conducted on patients treated with systemic corticosteroids during IPF admission. RESULTS In patients with IPF defined based on the ICD-10 (analysis 1), the RRs significantly decreased from March in 2020 to December 2021, except for June and September in 2020. Similarly, in patients with IPF defined based on the ICD-10 and RID (analysis 2), the RRs significantly decreased from March 2020 to December 2021, except for June and September 2020. In the sensitivity analysis of analysis 1, the RR significantly decreased in 2020 (0.93; 95%CI: 0.88-0.99; P = 0.029), whereas the RR in 2021 was not significantly different. The RRs in the sensitivity analysis of analysis 2 significantly decreased to 0.85 (0.79-0.92; P < 0.001) in 2020 and 0.82 (0.76-0.88; P < 0.001) in 2021. In the subgroup analysis, the rates of IPF admissions significantly decreased in 2020 and 2021 across both sexes, patients aged ≥ 60 years, and all household income groups. CONCLUSIONS The rate of IPF admissions significantly decreased during the COVID-19 pandemic. This result indicates that preventive measures against COVID-19 may effectively mitigate IPF exacerbation. Therefore, it is assumed that there is a close relationship between respiratory viral infections and IPF exacerbations.
Collapse
Affiliation(s)
- In-So Cho
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, 26426, Korea
| | - Jihye Lim
- Department of Medical Informatics and Biostatistics, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Min-Seok Chang
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, 26426, Korea
| | - Ji-Ho Lee
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju, 26426, Korea.
| |
Collapse
|
3
|
Qi J, Wu Y, Guo Z, Zhu S, Xiong J, Hu F, Liang X, Ye X. Fibroblast growth factor 21 alleviates idiopathic pulmonary fibrosis by inhibiting PI3K-AKT-mTOR signaling and stimulating autophagy. Int J Biol Macromol 2024; 273:132896. [PMID: 38851619 DOI: 10.1016/j.ijbiomac.2024.132896] [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: 05/12/2023] [Revised: 05/20/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive pulmonary disease with an unclear pathogenesis and no available specific drug treatment. The principal etiological factors are lung inflammation caused by environmental factors, damage to alveolar epithelial cells, leading to epithelial-mesenchymal transition (EMT), and the abnormal proliferation of fibroblasts. Here, we have demonstrated that fibroblast growth factor 21 (FGF21) ameliorates IPF via the autophagy pathway. We administered FGF21 to bleomycin (BLM)-treated mice, which ameliorated their defects in lung function, reduced the accumulation of collagen, restored tissue structure, reduced the deposition of hydroxyproline, reduced the expression of collagen I and α-SMA and increased the expression of E-cadherin. The expression of LC3BII and the number of autophagosomes were significantly higher in the lungs. The expression of AKT and mTOR was significantly reduced by FGF21 treatment. We also determined the effects of FGF21 in A549 cells treated with TGF-β, and found that FGF21 significantly inhibits activation of the AKT signaling pathway, thereby reducing TGF-β-induced EMT and preventing the uncontrolled proliferation of fibroblasts. We conclude that FGF21 ameliorates IPF by inhibiting the PI3K-AKT-mTOR signaling pathway and activating autophagy, which provides a theoretical basis for FGF21 to be used for the treatment of IPF.
Collapse
Affiliation(s)
- Jianying Qi
- School of chemical engineering, Jiangxi Normal University, 99 Ziyang Avenue, Nanchang 330022, China
| | - Yuanyuan Wu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Zhimou Guo
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Key Laboratory of Separation Science for Analytical Chemistry, Zhongshan Road 457, Dalian 116023, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jingjing Xiong
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Fei Hu
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Xinmiao Liang
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China; Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Key Laboratory of Separation Science for Analytical Chemistry, Zhongshan Road 457, Dalian 116023, China.
| | - Xianlong Ye
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China.
| |
Collapse
|
4
|
Toker Ç, Kuyucu Y, Şaker D, Kara S, Güzelel B, Mete UÖ. Investigation of miR-26b and miR-27b expressions and the effect of quercetin on fibrosis in experimental pulmonary fibrosis. J Mol Histol 2024; 55:25-35. [PMID: 37857923 DOI: 10.1007/s10735-023-10168-z] [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: 03/23/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023]
Abstract
In this study, investigation of the effects of Quercetin on Bleomycin-induced pulmonary fibrosis and fibrosis-associated molecules miR-26b and miR-27b was aimed. Control group was given 10% saline on the 0th day, and saline was administered for 21 days starting from the 8th day. Group 2 was given 50 mg/kg Quercetin for 21 days starting from the 8th day. Group 3 was given 10 mg/kg Bleomycin Sulfate on day 0, and sacrificed on the 22nd and 29th day. Group 4 was given 10 mg/kg Bleomycin Sulfate on the 0th day, and was given 50 mg/kg Quercetin for 14 days, and 21 days starting from day 8. Lung tissues were examined using light and electron microscopic, immunohistochemical and molecular biological methods. Injury groups revealed impaired alveolar structure, collagen accumulation and increased inflammatory cells in interalveolar septum. Fibrotic response was decreased and the alveolar structure was improved with Quercetin treatment. α-SMA expressions were higher in the injury groups, but lower in the treatment groups compared to the injury groups. E-cadherin expressions were decreased in the injury groups and showed stronger immunoreactivity in the treatment groups compared to the injury groups. miR-26b and miR-27b expressions were lower in the injury groups than the control groups, and higher in the treatment groups than the injury groups. Quercetin can be considered as a new treatment agent in the idiopathic pulmonary fibrosis, since it increases the expression levels of miR-26b and miR-27b which decrease in fibrosis, and has therapeutic effects on the histopathological changes.
Collapse
Affiliation(s)
- Çağrı Toker
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, 01330, Adana, Turkey
| | - Yurdun Kuyucu
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, 01330, Adana, Turkey.
| | - Dilek Şaker
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, 01330, Adana, Turkey
| | - Samet Kara
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, 01330, Adana, Turkey
| | - Bilge Güzelel
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, 01330, Adana, Turkey
| | - Ufuk Özgü Mete
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, 01330, Adana, Turkey
| |
Collapse
|
5
|
Scieszka D, Bolt AM, McCormick MA, Brigman JL, Campen MJ. Aging, longevity, and the role of environmental stressors: a focus on wildfire smoke and air quality. FRONTIERS IN TOXICOLOGY 2023; 5:1267667. [PMID: 37900096 PMCID: PMC10600394 DOI: 10.3389/ftox.2023.1267667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023] Open
Abstract
Aging is a complex biological process involving multiple interacting mechanisms and is being increasingly linked to environmental exposures such as wildfire smoke. In this review, we detail the hallmarks of aging, emphasizing the role of telomere attrition, cellular senescence, epigenetic alterations, proteostasis, genomic instability, and mitochondrial dysfunction, while also exploring integrative hallmarks - altered intercellular communication and stem cell exhaustion. Within each hallmark of aging, our review explores how environmental disasters like wildfires, and their resultant inhaled toxicants, interact with these aging mechanisms. The intersection between aging and environmental exposures, especially high-concentration insults from wildfires, remains under-studied. Preliminary evidence, from our group and others, suggests that inhaled wildfire smoke can accelerate markers of neurological aging and reduce learning capabilities. This is likely mediated by the augmentation of circulatory factors that compromise vascular and blood-brain barrier integrity, induce chronic neuroinflammation, and promote age-associated proteinopathy-related outcomes. Moreover, wildfire smoke may induce a reduced metabolic, senescent cellular phenotype. Future interventions could potentially leverage combined anti-inflammatory and NAD + boosting compounds to counter these effects. This review underscores the critical need to study the intricate interplay between environmental factors and the biological mechanisms of aging to pave the way for effective interventions.
Collapse
Affiliation(s)
- David Scieszka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Alicia M. Bolt
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Mark A. McCormick
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Jonathan L. Brigman
- Department of Neurosciences, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Matthew J. Campen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
6
|
Wang X, Deng X, Wu Y, Qian Z, Cai M, Li H, Lin H. Low-level ambient sulfur dioxide exposure and genetic susceptibility associated with incidence of idiopathic pulmonary fibrosis: A national prospective cohort study. CHEMOSPHERE 2023; 337:139362. [PMID: 37414299 DOI: 10.1016/j.chemosphere.2023.139362] [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: 03/07/2023] [Revised: 06/14/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND The association between long-term air pollution exposure and the development of idiopathic pulmonary fibrosis (IPF) has been established, but the evidence regarding the effect of low levels of air pollution, especially ambient sulfur dioxide (SO2), is limited. Besides, the combined effect and interaction between genetic susceptibility and ambient SO2 on IPF remain uncertain. METHODS This study retrieved data from 402,042 participants who were free of IPF at baseline in the UK Biobank. The annual mean concentration of ambient SO2 was estimated for each participant based on their residential addresses using a bilinear interpolation method. Cox proportional hazard models were used to examine the relationship between ambient SO2 and incident IPF. We further generated a polygenic risk score (PRS) for IPF and estimated the combined effects of genetic susceptibility and ambient SO2 on incident IPF. RESULTS After a median follow-up of 11.78 years, 2562 cases of IPF were identified. The results indicated that each 1 μg/m3 increase in ambient SO2 was associated with a hazard ratio (HR) (95% confidence interval [CI]) of 1.67 (1.58, 1.76) for incident IPF. The study found statistically significant synergistic additive interaction between genetic susceptibility and ambient SO2. Individuals with high genetic risk and high ambient SO2 exposure had a higher risk of developing IPF (HR = 7.48, 95% CI:5.66, 9.90). CONCLUSION The study suggests that long-term exposure to ambient SO2, even at concentrations lower than current air quality guidelines set by the Word Health Organization and European Union, may be an important risk factor for IPF. This risk is more pronounced among people with a high genetic risk. Therefore, these findings emphasize the need to consider the potential health effects of SO2 exposure and the necessity for stricter air quality standards.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, China
| | - Xu Deng
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, China
| | - Yinglin Wu
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, China
| | - Zhengmin Qian
- Department of Epidemiology and Biostatistics, College for Public Health and Social Justice, Saint Louis University, USA
| | - Miao Cai
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, China
| | - Haitao Li
- Department of Social Medicine and Health Service Management, Shenzhen University General Hospital, China
| | - Hualiang Lin
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, China.
| |
Collapse
|
7
|
Yang Y, Cao L, Xia Y, Li J. The effect of living environmental factors on cardiovascular diseases in Chinese adults: results from a cross-sectional and longitudinal study. Eur J Prev Cardiol 2023; 30:1063-1073. [PMID: 36537654 DOI: 10.1093/eurjpc/zwac304] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/01/2022] [Accepted: 12/16/2022] [Indexed: 08/23/2023]
Abstract
AIMS This study aimed to investigate the association between multiple living environmental factors and cardiovascular diseases (CVDs). METHODS AND RESULTS This study was conducted on the China Health and Retirement Longitudinal Study (CHARLS), with 12 489 subjects in the cross-sectional study and 7932 subjects in the 7-year follow-up. Living environmental factors included ambient fine particulate matter (PM2.5), indoor fuel use, tap water use, and residence type. Logistic regression and Cox proportional hazard regression models were applied to explore the association between living environmental risk factors and CVD events in a cross-sectional and a cohort analysis, respectively. Compared with subjects in the low-risk groups, those in the middle-risk (odd ratio [OR], 95% confidence interval [CI]: 1.203, 0.943-1.534) and high-risk groups (OR, 95% CI: 1.616, 1.259-2.074) showed increased risks of CVD prevalence when considering the combined effects of their living environment. During the follow-up, similar associations were observed (hazard ratio [HR], 1.541, 95% CI [1.142-2.080] for the high-risk group; HR 1.296, 95% CI [0.968-1.736] for the middle-risk group); P for trend = 0.003). CONCLUSION An overall poor living environmental quality is a potential risk factor for CVD. Future studies should focus more on the effects of exposure to multiple factors.
Collapse
Affiliation(s)
- Yao Yang
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 San Hao Street, Heping District, Shenyang 110004, Liaoning Province, China
| | - Limin Cao
- The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China
| | - Yang Xia
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, No. 36 San Hao Street, Heping District, Shenyang 110004, Liaoning Province, China
| | - Jian Li
- Department of Cardiovascular Medicine, The First Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shengyang 110001, Liaoning Province, China
| |
Collapse
|
8
|
Li Y, Du Z, Li T, Ren X, Yu Y, Duan J, Sun Z. MitoQ ameliorates PM 2.5-induced pulmonary fibrosis through regulating the mitochondria DNA homeostasis. CHEMOSPHERE 2023; 330:138745. [PMID: 37088202 DOI: 10.1016/j.chemosphere.2023.138745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/11/2023] [Accepted: 04/19/2023] [Indexed: 05/03/2023]
Abstract
Pulmonary fibrosis is a severe pulmonary disease, and may related to PM2.5 exposure. Our study aims to explore the pathogenesis of PM2.5-induced pulmonary fibrosis, and MitoQ protective effect in this process. Our results find that inflammatory cells aggregation and pulmonary fibrosis in mice lung after PM2.5 exposure. Moreover, Collagen I/III overproduction, EMT and TGF-β1/Smad2 pathway activation in mice lung and BEAS-2B after PM2.5 exposure. Fortunately, these changes were partially ameliorated after MitoQ treatment. Meanwhile, severe oxidative stress, mitochondrial homeostasis imbalance, overproduction of 8-oxoG (7,8-dihydro-8-oxoguanine), as well as the inhibition of SIRT3/OGG1 pathway have founded in mice lung or BEAS-2B after PM2.5 exposure, which were alleviated by MitoQ treatment. Collectively, our study found that oxidative stress, especially mitochondrial oxidative stress participates in the PM2.5-induced pulmonary fibrosis, and MitoQ intervention had a protective effect on this progress. Moreover, mitochondrial DNA homeostasis might participate in the pulmonary fibrosis caused by PM2.5 exposure. Our study provides a novel pathogenesis of PM2.5-caused pulmonary fibrosis and a possible targeted therapy for the pulmonary diseases triggered by PM2.5.
Collapse
Affiliation(s)
- Yang Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Zhou Du
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Tianyu Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Xiaoke Ren
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Yang Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
9
|
Yin L, Han K, Jiang B, Meng Q, Aschner M, Li X, Chen R. NAT10 accelerates pulmonary fibrosis through N4-acetylated TGFB1-initiated epithelial-to-mesenchymal transition upon ambient fine particulate matter exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 322:121149. [PMID: 36731737 DOI: 10.1016/j.envpol.2023.121149] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/29/2022] [Accepted: 01/23/2023] [Indexed: 06/18/2023]
Abstract
Exposure to ambient fine particulate matter (PM2.5) has been linked to a higher pulmonary fibrosis risk. Dysregulation of the epitranscriptome results in abnormal expression of mRNAs during fibrosis development. N4-acetylcytidine (ac4C) is one of the most frequent RNA epigenetic alterations, however, its function in PM2.5-triggered fibrosis is yet unknown. In this study, lung epithelial and murine models were established and exposed to PM2.5 to analyze the function of ac4C alteration in pulmonary fibrosis and underlying mechanisms. Meanwhile, the expression levels of only known ac4C "writer" protein, N-acetyltransferase 10 (NAT10), were significantly induced in pulmonary epithelia, relative to the control. Subsequently, NAT10 enhanced the stability of transforming growth factor beta 1 (TGFB1) mRNA as well as protein levels. As an up-stream driver, TGFB1 accelerated EMT and fibrosis process. Inhibition of NAT10 significantly protected against pulmonary EMT and fibrosis driven by PM2.5 exposure, whereas TGFB1 overexpression reversed the protective effects of NAT10 inhibition. Thus, NAT10 accelerated PM2.5-triggered pulmonary fibrosis via increasing TGFB1 mRNA stability in an ac4C-dependent manner. Our results reveal a pivotal role of NAT10-regulated mRNA ac4C acetylation in PM2.5-triggered pulmonary fibrosis and uncover the potential epitranscriptional mechanism.
Collapse
Affiliation(s)
- Lijia Yin
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Ke Han
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Bo Jiang
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Qingtao Meng
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY, 10461, USA
| | - Xiaobo Li
- School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Rui Chen
- School of Public Health, Capital Medical University, Beijing, 100069, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, 100069, China; Beijing laboratory of allergic diseases, Capital Medical University; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
10
|
Cui F, Sun Y, Xie J, Li D, Wu M, Song L, Hu Y, Tian Y. Air pollutants, genetic susceptibility and risk of incident idiopathic pulmonary fibrosis. Eur Respir J 2023; 61:13993003.00777-2022. [PMID: 36137588 DOI: 10.1183/13993003.00777-2022] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/06/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Air pollutants are considered as non-negligible risk factors of idiopathic pulmonary fibrosis (IPF). However, the relationship between long-term air pollution and the incidence of IPF is unknown. Our objective was to explore the associations of air pollutants with IPF risk and further assess the modification effect of genetic susceptibility. METHODS We used land-use regression model estimated concentrations of nitrogen dioxide (NO2), nitrogen oxides (NO x ) and particulate matter (fine particulate matter with diameter <2.5 μm (PM2.5) and particulate matter with diameter <10 μm (PM10)). The polygenic risk score (PRS) was constructed using 13 independent single nucleotide polymorphisms. Cox proportional hazard models were used to evaluate the associations of air pollutants with IPF risk and further investigate the modification effect of genetic susceptibility. Additionally, absolute risk was calculated. RESULTS Among 433 738 participants from the UK Biobank, the incidence of IPF was 27.45 per 100 000 person-years during a median follow-up of 11.78 years. The adjusted hazard ratios of IPF for each interquartile range increase in NO2, NO x and PM2.5 were 1.11 (95% CI 1.03-1.19), 1.07 (95% CI 1.01-1.13) and 1.09 (95% CI 1.02-1.17), respectively. PM2.5 had the highest population attribution risk, followed by NO x and NO2. There were additive interactions between NO2, NO x and PM2.5 and genetic susceptibility. Participants with a high PRS and high air pollution had the highest risk of incident IPF compared with those with a low PRS and low air pollution (adjusted hazard ratio: NO2 3.94 (95% CI 2.77-5.60), NO x 3.08 (95% CI 2.21-4.27), PM2.5 3.65 (95% CI 2.60-5.13) and PM10 3.23 (95% CI 2.32-4.50)). CONCLUSION Long-term exposures to air pollutants may elevate the risk of incident IPF. There are additive effects of air pollutants and genetic susceptibility on IPF risk.
Collapse
Affiliation(s)
- Feipeng Cui
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Feipeng Cui and Yu Sun contributed equally to this work
| | - Yu Sun
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Feipeng Cui and Yu Sun contributed equally to this work
| | - Junqing Xie
- Center for Statistics in Medicine, NDORMS, The Botnar Research Centre, University of Oxford, Oxford, UK
| | - Dankang Li
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Mingyang Wu
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Lulu Song
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yonghua Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Yaohua Tian
- Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Maternal and Child Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| |
Collapse
|
11
|
PM2.5 Exposure Induces Lung Injury and Fibrosis by Regulating Ferroptosis via TGF-β Signaling. DISEASE MARKERS 2022; 2022:7098463. [PMID: 36204510 PMCID: PMC9532166 DOI: 10.1155/2022/7098463] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/18/2022]
Abstract
Background. Lung fibrosis is a severe lung disorder featured by chronic nonspecific inflammation of the interstitial lung and deposition of collagen, leading to lung dysfunction. It has been identified that ferroptosis is involved in the progression of lung injury. Particulate matter (PM2.5) is reported to be correlated with the incidence of pulmonary fibrosis. However, mechanisms underlying ferroptosis in PM2.5-related lung fibrosis is unclear. In this study, we aimed to explore the effect of PM2.5 on ferroptosis in lung fibrosis and the related molecular mechanisms. Methods. PM2.5-treated mouse model and cell model were established. Fibrosis and tissue damage were measured by Masson’s trichrome staining and HE staining. Fibrosis biomarkers, such as α-SMA, collagen I, and collagen III, were examined by histological analysis. The ferroptosis phenotypes, including the levels of iron, Fe2+, MDA, and GSH, were measured by commercial kits. ROS generation was checked by DCFH-DA. The oxidative stress indicators, 3-nitro-L-tyrosine (3
-NT), 4-HNE, and protein carbonyl, were checked by enzyme linked immunosorbent assay (ELISA). The thiobarbituric acid reactive substances (TBARS) and GSH/GSSG ratio were assessed by TBARS assay kit and GSH/GSSG assay kit, respectively. TGF-β signaling was detected by Western blotting. Results. PM2.5 induced the lung injury and fibrosis in the mice model, along with elevated expression of fibrosis markers. PM2.5 enhanced oxidative stress in the lung of the mice. The SOD2 expression was reduced, and NRF2 expression was enhanced in the mice by the treatment with PM2.5. PM2.5 triggered ferroptosis, manifested as suppressed expression of GPX4 and SLC7A11, decreased levels of iron, Fe2+, and MDA, and increased GSH level in mouse model and cell model. The TGF-β and Smad3 signaling was inhibited by PM2.5. ROS inhibitor NAC reversed PM2.5-regulated ROS and ferroptosis in primary mouse lung epithelial cells. Conclusions. Therefore, we concluded that PM2.5 exposure induced lung injury and fibrosis by inducing ferroptosis via TGF-β signaling.
Collapse
|
12
|
Double Lung Transplantation for Idiopathic Pulmonary Fibrosis in a Patient with a History of Liver Transplantation and Prolonged Journey for Disease-Specific Antifibrotic Therapy. Case Rep Pulmonol 2022; 2022:4054339. [PMID: 35996614 PMCID: PMC9392625 DOI: 10.1155/2022/4054339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by uncontrolled progressive lung fibrosis with a median survival of 3 to 5 years. Although currently available pharmacotherapy cannot cure the disease, antifibrotics including pirfenidone and nintedanib were shown to slow disease progression and improve survival in IPF. Nevertheless, there is a knowledge gap on the safety of antifibrotics in patients after liver transplantation receiving concomitant immunosuppressive therapy. This case report of a 68-year-old male patient with IPF illustrates how a complex medical history has led to diagnostic and therapeutic challenges considerably affecting clinical decisions and impacting the patient's journey. The increasing severity of lung function impairment due to the progressive natural history of IPF ultimately led to severe respiratory failure. Double lung transplantation (LTx) was performed as the only therapeutic option in end-stage disease with the potential to improve quality of life and survival. To the best of our knowledge, this is the first case report describing the feasibility and safety of antifibrotic therapy with pirfenidone for IPF in a 68-year-old patient with a history of liver transplantation receiving concomitant immunosuppressive therapy with tacrolimus who underwent successful double lung transplantation when alternative medical interventions had been exhausted.
Collapse
|
13
|
Li Y, Shi T, Li X, Sun H, Xia X, Ji X, Zhang J, Liu M, Lin Y, Zhang R, Zheng Y, Tang J. Inhaled tire-wear microplastic particles induced pulmonary fibrotic injury via epithelial cytoskeleton rearrangement. ENVIRONMENT INTERNATIONAL 2022; 164:107257. [PMID: 35486965 DOI: 10.1016/j.envint.2022.107257] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 06/14/2023]
Abstract
Tire wear microplastic particles (TWMPs) are emerging microplastic pollutants that have gained increasing attention lately. However, the health effect of inhaled airborne TWMPs has never been explored before and may already be included in particulate matter morbidity and mortality. Here, we endeavored to address the preliminary study of TWMP inhalation-induced pulmonary toxic effects and its epigenetic mechanisms in C57BL/6 mice. As a result, restricted ventilatory dysfunction and fibrotic pathological changes were observed in TWMP-treaded mice. Further research found that attenuation of miR-1a-3p plays an important role in TWMP-induced lung injury. Results from in vitro study confirmed that cytoskeleton regulatory gene twinfilin-1 was one of the target genes of miR-1a-3p, and involved in cytoskeleton rearrangement caused by TWMP exposure. Mechanistically, miR-1a-3p inhibited the F-actin formation by targeting cytoskeletal regulatory proteins twinfilin-1, leading to TWMP-induced pulmonary fibrotic injury. While we are in the very early stages of explaining the role of epigenetics in TWMP-induced lung injury, the potential for the use of epigenetic marks as biomarkers is high and discoveries made in this field will likely bring us closer to better understanding this crucial mechanism.
Collapse
Affiliation(s)
- Yanting Li
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Teng Shi
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xin Li
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Huimin Sun
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaowen Xia
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Xiaoya Ji
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Jianzhong Zhang
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Meike Liu
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Yongfeng Lin
- School of Public Health, Qingdao University, Qingdao 266071, China
| | - Rong Zhang
- School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao 266071, China.
| | - Jinglong Tang
- School of Public Health, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
14
|
Majewski S, Szewczyk K, Jerczyńska H, Miłkowska-Dymanowska J, Białas AJ, Gwadera Ł, Piotrowski WJ. Longitudinal and Comparative Measures of Serum Chitotriosidase and YKL-40 in Patients With Idiopathic Pulmonary Fibrosis. Front Immunol 2022; 13:760776. [PMID: 35222369 PMCID: PMC8866556 DOI: 10.3389/fimmu.2022.760776] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
Background Although chitin is absent in humans, chitinases are present in healthy subjects and show dysregulated expression in a variety of diseases resulting from abnormal tissue injury and repair responses. It was shown that chitotriosidase (chitinase 1/CHIT1) and structurally-related chitinase 3-like 1 protein (CHI3L1/YKL-40) play important roles in the pathobiology of idiopathic pulmonary fibrosis (IPF), however little is known about their longitudinal serum levels and relationship to clinical measures in IPF. Methods The present study is the first to evaluate serial measurements of serum CHIT1 activity and YKL-40 concentrations in patients with IPF starting antifibrotic treatment and followed up for 24 months. In addition, baseline serum CHIT1 and YKL-40 were compared between patients with IPF and control subjects, and possible CHIT1 and YKL-40 relationships to longitudinal clinical assessments in IPF were explored. Results Baseline serum CHIT1 activity and YKL-40 concentrations were significantly elevated in patients with IPF compared to control subjects and showed similar discriminatory ability in distinguishing IPF from controls. No significant differences between the median serum CHIT1 activity and YKL-40 concentration measured over a study follow-up were noted. We found significantly elevated baseline serum CHIT1 activity in the progressors compared with the stables in the first year, while significantly increased baseline serum CHIT1 activity was noted in the stables compared to the progressors in the second year. Additionally, we observed a significant negative correlation between a change in serum YKL-40 concentration and a change in forced vital capacity (FVC) % predicted (% pred.) in the stables subgroup, whereas, a change in serum CHIT1 activity correlated negatively with a change in FVC% pred. in the progressors subgroup. Conclusions This explorative study findings add further evidence that CHIT1 and YKL-40 are upregulated in patients with IPF, and suggest that longitudinally stable serum CHIT1 activity and YKL-40 concentration levels may potentially be associated with the antifibrotic treatment response. In addition, our findings are supporting the possible role of CHIT1 and YKL-40 as candidate diagnostic and prognostic biomarkers in IPF. Further research is needed to validate present study findings.
Collapse
Affiliation(s)
| | - Karolina Szewczyk
- Department of Pathobiology of Respiratory Diseases, Medical University of Lodz, Lodz, Poland
| | - Hanna Jerczyńska
- Central Scientific Laboratory (CoreLab), Medical University of Lodz, Lodz, Poland
| | | | - Adam J Białas
- Department of Pathobiology of Respiratory Diseases, Medical University of Lodz, Lodz, Poland
| | - Łukasz Gwadera
- Department of Pneumology, Medical University of Lodz, Lodz, Poland
| | | |
Collapse
|
15
|
Treating Pulmonary Fibrosis with Non-Viral Gene Therapy: From Bench to Bedside. Pharmaceutics 2022; 14:pharmaceutics14040813. [PMID: 35456646 PMCID: PMC9027953 DOI: 10.3390/pharmaceutics14040813] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/30/2022] [Accepted: 04/02/2022] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive lung disease characterized by irreversible lung scarring, which achieves almost 80% five-year mortality rate. Undeniably, commercially available pharmaceuticals, such as pirfenidone and nintedanib, exhibit certain effects on improving the well-being of IPF patients, but the stubbornly high mortality still indicates a great urgency of developing superior therapeutics against this devastating disease. As an emerging strategy, gene therapy brings hope for the treatment of IPF by precisely regulating the expression of specific genes. However, traditional administration approaches based on viruses severely restrict the clinical application of gene therapy. Nowadays, non-viral vectors are raised as potential strategies for in vivo gene delivery, attributed to their low immunogenicity and excellent biocompatibility. Herein, we highlight a variety of non-viral vectors, such as liposomes, polymers, and proteins/peptides, which are employed in the treatment of IPF. By respectively clarifying the strengths and weaknesses of the above candidates, we would like to summarize the requisite features of vectors for PF gene therapy and provide novel perspectives on design-decisions of the subsequent vectors, hoping to accelerate the bench-to-bedside pace of non-viral gene therapy for IPF in clinical setting.
Collapse
|
16
|
Liang L, Cai Y, Lyu B, Zhang D, Chu S, Jing H, Rahimi K, Tong Z. Air pollution and hospitalization of patients with idiopathic pulmonary fibrosis in Beijing: a time-series study. Respir Res 2022; 23:81. [PMID: 35382829 PMCID: PMC8985349 DOI: 10.1186/s12931-022-01998-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
Background A small number of studies suggested that air pollution was associated with idiopathic pulmonary fibrosis (IPF) exacerbation, incidence and mortality. However, no studies to date were conducted in regions where air pollution is substantial. We aimed to investigate whether there are associations between acute increases in air pollution and hospitalization of patients with a confirmed primary diagnosis of IPF in Beijing. Methods Daily count of IPF hospitalizations (International Classification of Disease-10th Revision, J84.1) was obtained from an administrative database for 2013–2017 while daily city-wide average concentrations of PM10, PM2.5, NO2, Ozone, SO2 were obtained from 35 municipal monitoring stations for the same period. The association between daily IPF hospitalization and average concentration of each pollutant was analyzed with a generalized additive model estimating Poisson distribution. Results Daily 24-h mean PM2.5 concentration during 2013–2017 was 76.7 μg/m3. The relative risk (RR) of IPF hospitalization per interquartile range (IQR) higher (72 μg/m3) in PM2.5 was 1.049 (95% CI 1.024–1.074) and 1.031 (95% CI 1.007–1.056) for lag0 and moving averages 0–1 days respectively. No significant associations were observed for other lags. Statistically significant positive associations were also observed at lag0 with SO2, Ozone and NO2 (in men only). Positive associations were seen at moving averages 0–30 days for PM10 (RR per 86 μg/m3: 1.021, 95% CI 0.994–1.049), NO2 (RR per 30 μg/m3: 1.029, 95% CI 0.999–1.060), and SO2 (RR per 15 μg/m3: 1.060 (95% CI 1.025–1.097), but not with PM2.5 or Ozone. Conclusions Despite improvement in air quality since the implementation of clean air policy in 2013, acute exposure to higher levels of air pollution is significantly associated with IPF hospitalization in Beijing. Air quality policy should be continuously enforced to protect vulnerable IPF populations as well as the general public. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-01998-8.
Collapse
Affiliation(s)
- Lirong Liang
- Department of Clinical Epidemiology & Tobacco Dependence Treatment Research, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yutong Cai
- Centre for Environmental Health and Sustainability, Department of Health Sciences, University of Leicester, Leicester, LE1 7RH, UK
| | - Baolei Lyu
- Huayun Sounding Meteorology Technology Corporation, Beijing, China
| | - Di Zhang
- Department of Clinical Epidemiology & Tobacco Dependence Treatment Research, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Shuilian Chu
- Department of Clinical Epidemiology & Tobacco Dependence Treatment Research, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hang Jing
- Department of Clinical Epidemiology & Tobacco Dependence Treatment Research, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Kazem Rahimi
- Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Zhaohui Tong
- Department of Clinical Epidemiology & Tobacco Dependence Treatment Research, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China. .,Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
17
|
Nunes MC, Hale MJ, Mahtab S, Mabena FC, Dludlu N, Baillie VL, Thwala BN, Els T, du Plessis J, Laubscher M, Mckenzie S, Mtshali S, Menezes C, Serafin N, van Blydenstein S, Tsitsi M, Dulisse B, Madhi SA. Clinical characteristics and histopathology of COVID-19 related deaths in South African adults. PLoS One 2022; 17:e0262179. [PMID: 35051205 PMCID: PMC8775212 DOI: 10.1371/journal.pone.0262179] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/17/2021] [Indexed: 12/22/2022] Open
Abstract
Comparisons of histopathological features and microbiological findings between decedents with respiratory symptoms due to SARS-CoV-2 infection or other causes, in settings with high prevalence of HIV and Mycobacterium tuberculosis (MTB) infections have not been reported. Deaths associated with a positive ante-mortem SARS-CoV-2 PCR test and/or respiratory disease symptoms at Chris Hani Baragwanath Academic Hospital in Soweto, South Africa from 15th April to 2nd November 2020, during the first wave of the South African COVID-19 epidemic, were investigated. Deceased adult patients had post-mortem minimally-invasive tissue sampling (MITS) performed to investigate for SARS-CoV-2 infection and molecular detection of putative pathogens on blood and lung samples, and histopathology examination of lung, liver and heart tissue. During the study period MITS were done in patients displaying symptoms of respiratory disease including 75 COVID-19-related deaths (COVID+) and 42 non-COVID-19-related deaths (COVID-). The prevalence of HIV-infection was lower in COVID+ (27%) than in the COVID- (64%), MTB detection was also less common among COVID+ (3% vs 13%). Lung histopathology findings showed differences between COVID+ and COVID- in the severity of the morphological appearance of Type-II pneumocytes, alveolar injury and repair initiated by SARS-CoV-2 infection. In the liver necrotising granulomatous inflammation was more common among COVID+. No differences were found in heart analyses. The prevalence of bacterial co-infections was higher in COVID+. Most indicators of respiratory distress syndrome were undifferentiated between COVID+ and COVID- except for Type-II pneumocytes. HIV or MTB infection does not appear in these data to have a meaningful correspondence with COVID-related deaths.
Collapse
Affiliation(s)
- Marta C. Nunes
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Martin J. Hale
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Division of Anatomical Pathology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sana Mahtab
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fikile C. Mabena
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Paediatrics and Child Health, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Noluthando Dludlu
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Vicky L. Baillie
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Bukiwe N. Thwala
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Toyah Els
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jeanine du Plessis
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Marius Laubscher
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Shakeel Mckenzie
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sihle Mtshali
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Colin Menezes
- Department of Internal Medicine, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Natali Serafin
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sarah van Blydenstein
- Department of Internal Medicine, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Merika Tsitsi
- Department of Internal Medicine, Chris Hani Baragwanath Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Brian Dulisse
- BDII Analytics, Atlanta, GA, United States of America
| | - Shabir A. Madhi
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Technology/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
18
|
Hahad O, Kuntic M, Frenis K, Chowdhury S, Lelieveld J, Lieb K, Daiber A, Münzel T. Physical Activity in Polluted Air-Net Benefit or Harm to Cardiovascular Health? A Comprehensive Review. Antioxidants (Basel) 2021; 10:1787. [PMID: 34829658 PMCID: PMC8614825 DOI: 10.3390/antiox10111787] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 12/15/2022] Open
Abstract
Both exposure to higher levels of polluted air and physical inactivity are crucial risk factors for the development and progression of major noncommunicable diseases and, in particular, of cardiovascular disease. In this context, the World Health Organization estimated 4.2 and 3.2 million global deaths per year in response to ambient air pollution and insufficient physical activity, respectively. While regular physical activity is well known to improve general health, it may also increase the uptake and deposit of air pollutants in the lungs/airways and circulation, due to increased breathing frequency and minute ventilation, thus increasing the risk of cardiovascular disease. Thus, determining the tradeoff between the health benefits of physical activity and the potential harmful effects of increased exposure to air pollution during physical activity has important public health consequences. In the present comprehensive review, we analyzed evidence from human and animal studies on the combined effects of physical activity and air pollution on cardiovascular and other health outcomes. We further report on pathophysiological mechanisms underlying air pollution exposure, as well as the protective effects of physical activity with a focus on oxidative stress and inflammation. Lastly, we provide mitigation strategies and practical recommendations for physical activity in areas with polluted air.
Collapse
Affiliation(s)
- Omar Hahad
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.H.); (M.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany;
| | - Marin Kuntic
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.H.); (M.K.)
| | - Katie Frenis
- Department of Hematology/Oncology, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Sourangsu Chowdhury
- Atmospheric Chemistry Department, Max Planck Institute for Chemistry, 55122 Mainz, Germany; (S.C.); (J.L.)
| | - Jos Lelieveld
- Atmospheric Chemistry Department, Max Planck Institute for Chemistry, 55122 Mainz, Germany; (S.C.); (J.L.)
- Climate and Atmosphere Research Center, The Cyprus Institute, Nicosia 2121, Cyprus
| | - Klaus Lieb
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany;
- Department of Psychiatry and Psychotherapy, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.H.); (M.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology—Cardiology I, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (O.H.); (M.K.)
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany
| |
Collapse
|
19
|
Kojima T, Shindo Y, Konno T, Kodera Y, Arai W, Miyakawa M, Ohwada K, Tanaka H, Tsujiwaki M, Sakuma Y, Kikuchi S, Ohkuni T, Takano K, Watanabe A, Kohno T. Dysfunction of epithelial permeability barrier induced by HMGB1 in 2.5D cultures of human epithelial cells. Tissue Barriers 2021; 10:1972760. [PMID: 34538217 DOI: 10.1080/21688370.2021.1972760] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Airway and intestinal epithelial permeability barriers are crucial in epithelial homeostasis. High mobility group box 1 (HMGB1), increased by various stimuli, is involved in the induction of airway inflammation, as well as the pathogenesis of inflammatory bowel disease. HMGB1 enhances epithelial hyperpermeability. Two-and-a-half dimensional (2.5D) culture assays are experimentally convenient and induce cells to form a more physiological tissue architecture than 2D culture assays for molecular transfer mechanism analysis. In 2.5D culture, treatment with HMGB1 induced permeability of FITC-dextran into the lumen formed by human lung, nasal and intestinal epithelial cells. The tricellular tight junction molecule angulin-1/LSR is responsible for the epithelial permeability barrier at tricellular contacts and contributes to various human airway and intestinal inflammatory diseases. In this review, we indicate the mechanisms including angulin-1/LSR and multiple signaling in dysfunction of the epithelial permeability barrier induced by HMGB1 in 2.5D culture of human airway and intestinal epithelial cells.
Collapse
Affiliation(s)
- Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuma Shindo
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Thoracic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuki Kodera
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Wataru Arai
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Thoracic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Maki Miyakawa
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,IBD Hospital, Sapporo, Japan
| | - Kizuku Ohwada
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | | | - Mitsuhiro Tsujiwaki
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuji Sakuma
- Department of Molecular Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shin Kikuchi
- Department of Anatomy, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tsuyoshi Ohkuni
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenichi Takano
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Atsushi Watanabe
- Department of Thoracic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
20
|
Wu X, Huang J, Wang J, Xu Y, Yang X, Sun M, Shi J. Multi-Pharmaceutical Activities of Chinese Herbal Polysaccharides in the Treatment of Pulmonary Fibrosis: Concept and Future Prospects. Front Pharmacol 2021; 12:707491. [PMID: 34489700 PMCID: PMC8418122 DOI: 10.3389/fphar.2021.707491] [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: 05/10/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022] Open
Abstract
Pulmonary fibrosis is a fatal chronic progressive respiratory disease, characterized by continuous scarring of the lung parenchyma, leading to respiratory failure and death. The incidence of PF has increased over time. There are drugs, yet, there are some limitations. Hence, it is of importance to find new therapies and new drugs to replace the treatment of pulmonary fibrosis. In recent years, there have been a great number of research reports on the treatment of traditional Chinese medicine polysaccharides in various system fields. Among them, the treatment of PF has also gained extensive attention. This review summarized the source of polysaccharides, the drug activity of traditional Chinese medicine, and the protective effects on targets of Pulmonary fibrosis. We hope it can inspire researchers to design and develop polysaccharides, serving as a reference for potential clinical therapeutic drugs.
Collapse
Affiliation(s)
- Xianbo Wu
- School of Sports Medicine and Health, Chegdu Sport University, Chengdu, China
| | - Jianli Huang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jie Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yihua Xu
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinwei Yang
- School of Sports Medicine and Health, Chegdu Sport University, Chengdu, China
| | - Minghan Sun
- Central of Reproductive Medicine, Department of Obstetrics and Gynecology, School of Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
21
|
Han X, Ding C, Sang X, Peng M, Yang Q, Ning Y, Lv Q, Shan Q, Hao M, Wang K, Wu X, Zhang H, Cao G. Targeting Sirtuin1 to treat aging-related tissue fibrosis: From prevention to therapy. Pharmacol Ther 2021; 229:107983. [PMID: 34480962 DOI: 10.1016/j.pharmthera.2021.107983] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 12/19/2022]
Abstract
Fibrosis, which is characterized by excessive extracellular matrix (ECM) deposition, is a wound-healing response to organ injury and may promote cancer and failure in various organs, such as the heart, liver, lung, and kidney. Aging associated with oxidative stress and inflammation exacerbates cellular dysfunction, tissue failure, and body function disorders, all of which are closely related to fibrosis. Sirtuin-1 (SIRT1) is a class III histone deacetylase that regulates growth, transcription, aging, and metabolism in various organs. This protein is downregulated in organ injury and fibrosis associated with aging. Its expression and distribution change with age in different organs and play critical roles in tissue oxidative stress and inflammation. This review first described the background on fibrosis and regulatory functions of SIRT1. Second, we summarized the relationships of SIRT1 with other proteins and its protective action during fibrosis in the heart, liver, lung and kidney. Third, the activation of SIRT1 in therapies of tissue fibrosis, especially in liver fibrosis and aging-related tissue injury, was analyzed. In conclusion, SIRT1 targeting may be a new therapeutic strategy in fibrosis.
Collapse
Affiliation(s)
- Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chuan Ding
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - XiaNan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - MengYun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Ning
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Lv
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - QiYuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Min Hao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - KuiLong Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Wu
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hongyan Zhang
- Cancer Hospital of The University of Chinese Academy of Sciences, Hangzhou, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
22
|
Serial Measurements of Circulating KL-6, SP-D, MMP-7, CA19-9, CA-125, CCL18, and Periostin in Patients with Idiopathic Pulmonary Fibrosis Receiving Antifibrotic Therapy: An Exploratory Study. J Clin Med 2021; 10:jcm10173864. [PMID: 34501312 PMCID: PMC8432145 DOI: 10.3390/jcm10173864] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/10/2021] [Accepted: 08/26/2021] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and inevitably fatal disease with a heterogeneous clinical course. This study aimed to evaluate the usefulness of circulating biomarkers in routine IPF clinical practice. We conducted an exploratory study in a cohort of 28 IPF subjects qualified for anti-fibrotic therapy with up to 24 months serial measurements of seven IPF biomarkers, including those that are well-established, Krebs von den Lungen-6 (KL-6), surfactant protein D (SP-D), matrix metalloproteinase 7 (MMP-7), and more recently introduced ones, cancer antigen 19-9 (CA19-9), cancer antigen 125 (CA-125), chemokine (C-C motif) ligand 18 (CCL18), and periostin. Among studied biomarkers, SP-D had the highest diagnostic accuracy to differentiate IPF subjects from controls, followed by MMP-7 and KL-6. At each study timepoint, KL-6 levels correlated inversely with forced vital capacity % predicted (FVC% pred.), and transfer factor of the lung for carbon monoxide % predicted (TL,CO% pred.), while SP-D levels correlated inversely with FVC% pred. and TL,CO% pred. at 24 months of anti-fibrotic therapy. Baseline KL-6 and CA19-9 concentrations were significantly elevated in patients with progressive disease in comparison to patients with stable disease. In addition, in the progressors subgroup CA19-9 concentrations significantly increased over the second year of study follow-up. In patients with progressive disease, we observed a significant inverse correlation between a change in SP-D levels and a change in FVC% pred. in the first year of treatment, whereas in the second year a significant inverse correlation between a change in KL-6 levels and a change in FVC% pred. was noted. Our study findings support the view that both well-established IPF biomarkers, including KL-6, SP-D, and MMP-7, and more recently introduced ones, like CA19-9, have the potential to support clinical practice in IPF.
Collapse
|
23
|
He Z, Peng H, Gao M, Liang G, Zeng M, Zhang X. p300/Sp1-Mediated High Expression of p16 Promotes Endothelial Progenitor Cell Senescence Leading to the Occurrence of Chronic Obstructive Pulmonary Disease. Mediators Inflamm 2021; 2021:5599364. [PMID: 34456628 PMCID: PMC8397552 DOI: 10.1155/2021/5599364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/19/2021] [Accepted: 08/02/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Chronic obstructive pulmonary disease (COPD) is a common chronic disease and develops rapidly into a grave public health problem worldwide. However, what exactly causes the occurrence of COPD remains largely unclear. Here, we are trying to explore whether the high expression of p16 mediated by p300/Sp1 can cause chronic obstructive pulmonary disease through promoting the senescence of endothelial progenitor cells (EPCs). METHODS Peripheral blood EPCs were isolated from nonsmoking non-COPD, smoking non-COPD, and smoking COPD patients. The expressions of p16, p300, and senescence-related genes were detected by RT-PCR and Western Blot. Then, we knocked down or overexpressed Sp1 and p300 and used the ChIP assay to detect the histone H4 acetylation level in the promoter region of p16, CCK8 to detect cell proliferation, flow cytometry to detect the cell cycle, and β-galactosidase staining to count the proportion of senescent cells. RESULTS The high expression of p16 was found in peripheral blood EPCs of COPD patients; the cigarette smoke extract (CSE) led to the increase of p16. The high expression of p16 in EPCs promoted cell cycle arrest and apoptosis. The CSE-mediated high expression of p16 promoted cell senescence. The expression of p300 was increased in peripheral blood EPCs of COPD patients. Moreover, p300/Sp1 enhanced the histone H4 acetylation level in the promoter region of p16, thereby mediating the senescence of EPCs. And knockdown of p300/Sp1 could rescue CSE-mediated cell senescence. CONCLUSION p300/Sp1 enhanced the histone H4 acetylation level in the p16 promoter region to mediate the senescence of EPCs.
Collapse
Affiliation(s)
- Zhihui He
- Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, China
| | - Huaihuai Peng
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan, China
| | - Min Gao
- Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, China
| | - Guibin Liang
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan, China
| | - Menghao Zeng
- Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, China
| | - Xuefeng Zhang
- Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, 410013 Hunan, China
| |
Collapse
|