1
|
Xu M, Feng P, Yan J, Li L. Mitochondrial quality control: a pathophysiological mechanism and potential therapeutic target for chronic obstructive pulmonary disease. Front Pharmacol 2025; 15:1474310. [PMID: 39830343 PMCID: PMC11739169 DOI: 10.3389/fphar.2024.1474310] [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: 08/01/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent chronic respiratory disease worldwide. Mitochondrial quality control mechanisms encompass processes such as mitochondrial biogenesis, fusion, fission, and autophagy, which collectively maintain the quantity, morphology, and function of mitochondria, ensuring cellular energy supply and the progression of normal physiological activities. However, in COPD, due to the persistent stimulation of harmful factors such as smoking and air pollution, mitochondrial quality control mechanisms often become deregulated, leading to mitochondrial dysfunction. Mitochondrial dysfunction plays a pivotal role in the pathogenesis of COPD, contributing toinflammatory response, oxidative stress, cellular senescence. However, therapeutic strategies targeting mitochondria remain underexplored. This review highlights recent advances in mitochondrial dysfunction in COPD, focusing on the role of mitochondrial quality control mechanisms and their dysregulation in disease progression. We emphasize the significance of mitochondria in the pathophysiological processes of COPD and explore potential strategies to regulate mitochondrial quality and improve mitochondrial function through mitochondrial interventions, aiming to treat COPD effectively. Additionally, we analyze the limitations and challenges of existing therapeutic strategies, aiming to provide new insights and methods for COPD treatment.
Collapse
Affiliation(s)
- Mengjiao Xu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng Feng
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Ferguson Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jun Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
2
|
Li D, Zhang X, Song Z, Zhao S, Huang Y, Qian W, Cai X. Advances in common in vitro cellular models of pulmonary fibrosis. Immunol Cell Biol 2024; 102:557-569. [PMID: 38714318 DOI: 10.1111/imcb.12756] [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: 08/12/2023] [Revised: 12/24/2023] [Accepted: 04/09/2024] [Indexed: 05/09/2024]
Abstract
The development of in vitro models is essential for a comprehensive understanding and investigation of pulmonary fibrosis (PF) at both cellular and molecular levels. This study presents a literature review and an analysis of various cellular models used in scientific studies, specifically focusing on their applications in elucidating the pathogenesis of PF. Our study highlights the importance of taking a comprehensive approach to studing PF, emphasizing the necessity of considering multiple cell types and organs and integrating diverse analytical perspectives. Notably, primary cells demonstrate remarkable cell growth characteristics and gene expression profiles; however, their limited availability, maintenance challenges, inability for continuous propagation and susceptibility to phenotypic changes over time significantly limit their utility in scientific investigation. By contrast, immortalized cell lines are easily accessible, cultured and continuously propagated, although they may have some phenotypic differences from primary cells. Furthermore, in vitro coculture models offer a more practical and precise method to explore complex interactions among cells, tissues and organs. Consequently, when developing models of PF, researchers should thoroughly assess the advantages, limitations and relevant mechanisms of different cell models to ensure their selection is consistent with the research objectives.
Collapse
Affiliation(s)
- Die Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinyue Zhang
- Department of Lung Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ziqiong Song
- Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Academy of Occupational Health and Occupational Medicine, Jinan, Shandong, China
| | - Shan Zhao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuan Huang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Weibin Qian
- Department of Lung Disease, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinrui Cai
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
3
|
Zhao Y, Wu Z. TROP2 promotes PINK1-mediated mitophagy and apoptosis to accelerate the progression of senile chronic obstructive pulmonary disease by up-regulating DRP1 expression. Exp Gerontol 2024; 191:112441. [PMID: 38685507 DOI: 10.1016/j.exger.2024.112441] [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: 01/11/2024] [Revised: 04/06/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic airway inflammatory disease characterised by irreversible airflow limitation. The elderly are a vulnerable population for developing COPD. With the growth of age, physiological degenerative changes occur in the thorax, bronchus, lung and vascular wall, which can lead to age-related physiological attenuation of lung function in the elderly, so the prevalence of COPD increases with age. Its pathogenesis has not yet been truly clarified. Mitophagy plays an important role in maintaining the stability of mitochondrial function and intracellular environment by scavenging damaged mitochondria. Currently, studies have shown that trophoblast antigen 2 (TROP2) expression is up-regulated in airway basal cells of patients with COPD, suggesting that TROP2 is involved in the progression of COPD. However, whether it is involved in disease progression by regulating mitochondrial function remains unclear. In this study, compared with non-smoking non-COPD patients, the expression of TROP2 in lung tissues of smoking non-COPD patients and patients with COPD increased, and TROP2 expression in patients with COPD was higher than that in smoking non-COPD patients. To further explore the role of TROP2, we stimulated BEAS-2B with cigarette smoke to construct an in vitro model. We found that TROP2 expression increased, whereas TROP2 silencing reversed the cigarette smoke extract-induced decrease in mitochondrial membrane potential, increased reactive oxygen species content, decreased adenosine triphosphate (ATP) production, increased inflammatory factor secretion and increased apoptosis. In addition, we searched online bioinformatics and screened the gene dynamin-related protein 1 (DRP1) related to mitophagy as the research object. Co-IP assay verified the binding relationship between DRP1 and TROP2. Further study found that TROP2 promoted mitophagy and apoptosis of BEAS-2B cells by up-regulating the expression of DRP1. In addition, PTEN-induced putative kinase 1 (PINK1) is a potential binding protein of DRP1, and DRP1 accelerated mitophagy and apoptosis of BEAS-2B cells by promoting the expression of PINK1. We established a COPD SD rat model by cigarette smoke exposure and LPS instillation and treated it by intraperitoneal injection of si-TROP2. The results showed that TROP2 silencing restored lung function and reduced the secretion of inflammatory factors in bronchoalveolar lavage fluid. In conclusion, TROP2 can be used as a new reference for COPD treatment.
Collapse
Affiliation(s)
- Yipu Zhao
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Zhengjie Wu
- Shandong Public Health Clinical Center, Shandong University, Jinan 250013, Shandong, China.
| |
Collapse
|
4
|
He Q, Li P, Han L, Yang C, Jiang M, Wang Y, Han X, Cao Y, Liu X, Wu W. Revisiting airway epithelial dysfunction and mechanisms in chronic obstructive pulmonary disease: the role of mitochondrial damage. Am J Physiol Lung Cell Mol Physiol 2024; 326:L754-L769. [PMID: 38625125 DOI: 10.1152/ajplung.00362.2023] [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: 11/22/2023] [Revised: 03/20/2024] [Accepted: 04/10/2024] [Indexed: 04/17/2024] Open
Abstract
Chronic exposure to environmental hazards causes airway epithelial dysfunction, primarily impaired physical barriers, immune dysfunction, and repair or regeneration. Impairment of airway epithelial function subsequently leads to exaggerated airway inflammation and remodeling, the main features of chronic obstructive pulmonary disease (COPD). Mitochondrial damage has been identified as one of the mechanisms of airway abnormalities in COPD, which is closely related to airway inflammation and airflow limitation. In this review, we evaluate updated evidence for airway epithelial mitochondrial damage in COPD and focus on the role of mitochondrial damage in airway epithelial dysfunction. In addition, the possible mechanism of airway epithelial dysfunction mediated by mitochondrial damage is discussed in detail, and recent strategies related to airway epithelial-targeted mitochondrial therapy are summarized. Results have shown that dysregulation of mitochondrial quality and oxidative stress may lead to airway epithelial dysfunction in COPD. This may result from mitochondrial damage as a central organelle mediating abnormalities in cellular metabolism. Mitochondrial damage mediates procellular senescence effects due to mitochondrial reactive oxygen species, which effectively exacerbate different types of programmed cell death, participate in lipid metabolism abnormalities, and ultimately promote airway epithelial dysfunction and trigger COPD airway abnormalities. These can be prevented by targeting mitochondrial damage factors and mitochondrial transfer. Thus, because mitochondrial damage is involved in COPD progression as a central factor of homeostatic imbalance in airway epithelial cells, it may be a novel target for therapeutic intervention to restore airway epithelial integrity and function in COPD.
Collapse
Affiliation(s)
- Qinglan He
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Peijun Li
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lihua Han
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Chen Yang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Meiling Jiang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yingqi Wang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoyu Han
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Yuanyuan Cao
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiaodan Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weibing Wu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
5
|
Tulen CBM, van de Wetering C, Schiffers CHJ, Weltjens E, Benedikter BJ, Leermakers PA, Boukhaled JH, Drittij MJ, Schmeck BT, Reynaert NL, Opperhuizen A, van Schooten FJ, Remels AHV. Alterations in the molecular control of mitochondrial turnover in COPD lung and airway epithelial cells. Sci Rep 2024; 14:4821. [PMID: 38413800 PMCID: PMC10899608 DOI: 10.1038/s41598-024-55335-8] [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: 03/01/2023] [Accepted: 02/22/2024] [Indexed: 02/29/2024] Open
Abstract
Abnormal mitochondria have been observed in bronchial- and alveolar epithelial cells of patients with chronic obstructive pulmonary disease (COPD). However, it is unknown if alterations in the molecular pathways regulating mitochondrial turnover (mitochondrial biogenesis vs mitophagy) are involved. Therefore, in this study, the abundance of key molecules controlling mitochondrial turnover were assessed in peripheral lung tissue from non-COPD patients (n = 6) and COPD patients (n = 11; GOLDII n = 4/11; GOLDIV n = 7/11) and in both undifferentiated and differentiated human primary bronchial epithelial cells (PBEC) from non-COPD patients and COPD patients (n = 4-7 patients/group). We observed significantly decreased transcript levels of key molecules controlling mitochondrial biogenesis (PPARGC1B, PPRC1, PPARD) in peripheral lung tissue from severe COPD patients. Interestingly, mRNA levels of the transcription factor TFAM (mitochondrial biogenesis) and BNIP3L (mitophagy) were increased in these patients. In general, these alterations were not recapitulated in undifferentiated and differentiated PBECs with the exception of decreased PPARGC1B expression in both PBEC models. Although these findings provide valuable insight in these pathways in bronchial epithelial cells and peripheral lung tissue of COPD patients, whether or not these alterations contribute to COPD pathogenesis, underlie changes in mitochondrial function or may represent compensatory mechanisms remains to be established.
Collapse
Affiliation(s)
- Christy B M Tulen
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Pharmacology and Toxicology, Maastricht University Medical Center+, Universiteitssingel 50, 6629 ER, Maastricht, The Netherlands
| | - Cheryl van de Wetering
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Respiratory Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Caspar H J Schiffers
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Respiratory Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ellen Weltjens
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Pharmacology and Toxicology, Maastricht University Medical Center+, Universiteitssingel 50, 6629 ER, Maastricht, The Netherlands
| | - Birke J Benedikter
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Microbiology, Maastricht University Medical Center, Maastricht, The Netherlands
- Institute for Lung Research, Philipps-University Marburg, Marburg, Germany
- Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Pieter A Leermakers
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Pharmacology and Toxicology, Maastricht University Medical Center+, Universiteitssingel 50, 6629 ER, Maastricht, The Netherlands
| | - Juliana H Boukhaled
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Pharmacology and Toxicology, Maastricht University Medical Center+, Universiteitssingel 50, 6629 ER, Maastricht, The Netherlands
| | - Marie-José Drittij
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Pharmacology and Toxicology, Maastricht University Medical Center+, Universiteitssingel 50, 6629 ER, Maastricht, The Netherlands
| | - Bernd T Schmeck
- Institute for Lung Research, Philipps-University Marburg, Marburg, Germany
- Department for Respiratory and Critical Care Medicine, Clinic for Respiratory Infections, University Medical Center Marburg, Marburg, Germany
- German Centers for Lung Research (DZL) and for Infectious Disease Research (DZIF), SYNMIKRO Center for Synthetic Microbiology, Philipps-University Marburg, 35037, Marburg, Germany
- Member of the German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Niki L Reynaert
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Respiratory Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Primary Lung Culture (PLUC) Facility, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Antoon Opperhuizen
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Pharmacology and Toxicology, Maastricht University Medical Center+, Universiteitssingel 50, 6629 ER, Maastricht, The Netherlands
- Office of Risk Assessment and Research, Netherlands Food and Consumer Product Safety Authority (NVWA), Utrecht, The Netherlands
| | - Frederik-Jan van Schooten
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Pharmacology and Toxicology, Maastricht University Medical Center+, Universiteitssingel 50, 6629 ER, Maastricht, The Netherlands
| | - Alexander H V Remels
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Department of Pharmacology and Toxicology, Maastricht University Medical Center+, Universiteitssingel 50, 6629 ER, Maastricht, The Netherlands.
| |
Collapse
|
6
|
Liu YB, Hong JR, Jiang N, Jin L, Zhong WJ, Zhang CY, Yang HH, Duan JX, Zhou Y. The Role of Mitochondrial Quality Control in Chronic Obstructive Pulmonary Disease. J Transl Med 2024; 104:100307. [PMID: 38104865 DOI: 10.1016/j.labinv.2023.100307] [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: 06/28/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a major cause of morbidity, mortality, and health care use worldwide with heterogeneous pathogenesis. Mitochondria, the powerhouses of cells responsible for oxidative phosphorylation and energy production, play essential roles in intracellular material metabolism, natural immunity, and cell death regulation. Therefore, it is crucial to address the urgent need for fine-tuning the regulation of mitochondrial quality to combat COPD effectively. Mitochondrial quality control (MQC) mainly refers to the selective removal of damaged or aging mitochondria and the generation of new mitochondria, which involves mitochondrial biogenesis, mitochondrial dynamics, mitophagy, etc. Mounting evidence suggests that mitochondrial dysfunction is a crucial contributor to the development and progression of COPD. This article mainly reviews the effects of MQC on COPD as well as their specific regulatory mechanisms. Finally, the therapeutic approaches of COPD via MQC are also illustrated.
Collapse
Affiliation(s)
- Yu-Biao Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jie-Ru Hong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Nan Jiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Ling Jin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jia-Xi Duan
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yong Zhou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China.
| |
Collapse
|
7
|
Liu L, Li Y, Chen G, Chen Q. Crosstalk between mitochondrial biogenesis and mitophagy to maintain mitochondrial homeostasis. J Biomed Sci 2023; 30:86. [PMID: 37821940 PMCID: PMC10568841 DOI: 10.1186/s12929-023-00975-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023] Open
Abstract
Mitochondrial mass and quality are tightly regulated by two essential and opposing mechanisms, mitochondrial biogenesis (mitobiogenesis) and mitophagy, in response to cellular energy needs and other cellular and environmental cues. Great strides have been made to uncover key regulators of these complex processes. Emerging evidence has shown that there exists a tight coordination between mitophagy and mitobiogenesis, and their defects may cause many human diseases. In this review, we will first summarize the recent advances made in the discovery of molecular regulations of mitobiogenesis and mitophagy and then focus on the mechanism and signaling pathways involved in the simultaneous regulation of mitobiogenesis and mitophagy in the response of tissue or cultured cells to energy needs, stress, or pathophysiological conditions. Further studies of the crosstalk of these two opposing processes at the molecular level will provide a better understanding of how the cell maintains optimal cellular fitness and function under physiological and pathophysiological conditions, which holds promise for fighting aging and aging-related diseases.
Collapse
Affiliation(s)
- Lei Liu
- Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| | - Yanjun Li
- Center of Cell Response, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Guo Chen
- Center of Cell Response, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Quan Chen
- Center of Cell Response, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
8
|
You Y, Wang H, Wang Q, Yu Z, Zhao Z, Zhuang L, Zeng S, Zheng J, Wen W. Silencing USP19 alleviates cigarette smoke extract-induced mitochondrial dysfunction in BEAS-2B cells by targeting FUNDC1. Open Med (Wars) 2023; 18:20230798. [PMID: 37808166 PMCID: PMC10560033 DOI: 10.1515/med-2023-0798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/31/2023] [Accepted: 08/17/2023] [Indexed: 10/10/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is commonly caused by smoking. FUN14 domain-containing protein 1 (FUNDC1) plays a fundamental role in mitochondrial autophagy and apoptosis in cigarette smoke extract (CSE)-treated BEAS-2B cells. The present study investigated the mechanism of action of FUNDC1 in mitochondrial dysfunction and apoptosis in CSE-treated BEAS-2B cells. The interaction between ubiquitin-specific peptidase 19 (USP19) and FUNDC1 was analyzed using co-immunoprecipitation. Effects of USP19 knockdown and/or FUNDC1 overexpression on the survival, apoptosis, mitochondrial membrane potential, and oxygen consumption rate (OCR) of BEAS-2B cells treated with 15% CSE were determined. In BEAS-2B cells, CSE inhibited cell survival, promoted apoptosis, increased the expression of USP19 and FUNDC1, increased the ratio of LC3 II to LC3 I (LC3 II/I), and decreased mitochondrial membrane potential and TOM20 levels. In CSE-treated BEAS-2B cells, USP19 knockdown reduced FUNDC1 and LC3 II/I, increased the levels of TOM20, improved cell survival, mitochondrial membrane potential, and OCR, and inhibited apoptosis. USP19 deubiquitinates FUNDC1. FUNDC1 overexpression inhibited the effect of USP19 knockdown in CSE-treated BEAS-2B cells. Overall, decreasing USP19 expression alleviates CSE-induced mitochondrial dysfunction in BEAS-2B cells by downregulating FUNDC1, providing novel insights into the molecular mechanism of FUNDC1 regulation in COPD.
Collapse
Affiliation(s)
- Yanjing You
- Department of Respiratory and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, 900TH Hospital of Joint Logistics Support Force, PLA, Fuzhou 350025, Fujian, P.R. China
| | - Huijuan Wang
- Graduate College of Fujian Medical University, Fuzhou350025, China
| | - Qing Wang
- Department of Respiratory and Critical Care Medicine, The Third Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou350108, Fujian, China
| | - Zongyang Yu
- Department of Respiratory and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, 900TH Hospital of Joint Logistics Support Force, PLA, Fuzhou 350025, Fujian, P.R. China
| | - Zhongquan Zhao
- Department of Respiratory and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, 900TH Hospital of Joint Logistics Support Force, PLA, Fuzhou 350025, Fujian, P.R. China
| | - Liying Zhuang
- Graduate College of Fujian Medical University, Fuzhou350025, China
| | - Shengyuan Zeng
- Graduate College of Fujian Medical University, Fuzhou350025, China
| | - Jinyang Zheng
- Graduate College of Fujian Medical University, Fuzhou350025, China
| | - Wen Wen
- Department of Respiratory and Critical Care Medicine, Fuzhou General Hospital of Fujian Medical University, Dongfang Hospital of Xiamen University, 900TH Hospital of Joint Logistics Support Force, PLA, No. 156, Xi’erhuan North Road, Gulou District, Fuzhou 350025, Fujian, P.R. China
| |
Collapse
|
9
|
Albano GD, Montalbano AM, Gagliardo R, Profita M. Autophagy/Mitophagy in Airway Diseases: Impact of Oxidative Stress on Epithelial Cells. Biomolecules 2023; 13:1217. [PMID: 37627282 PMCID: PMC10452925 DOI: 10.3390/biom13081217] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Autophagy is the key process by which the cell degrades parts of itself within the lysosomes. It maintains cell survival and homeostasis by removing molecules (particularly proteins), subcellular organelles, damaged cytoplasmic macromolecules, and by recycling the degradation products. The selective removal or degradation of mitochondria is a particular type of autophagy called mitophagy. Various forms of cellular stress (oxidative stress (OS), hypoxia, pathogen infections) affect autophagy by inducing free radicals and reactive oxygen species (ROS) formation to promote the antioxidant response. Dysfunctional mechanisms of autophagy have been found in different respiratory diseases such as chronic obstructive lung disease (COPD) and asthma, involving epithelial cells. Several existing clinically approved drugs may modulate autophagy to varying extents. However, these drugs are nonspecific and not currently utilized to manipulate autophagy in airway diseases. In this review, we provide an overview of different autophagic pathways with particular attention on the dysfunctional mechanisms of autophagy in the epithelial cells during asthma and COPD. Our aim is to further deepen and disclose the research in this direction to stimulate the develop of new and selective drugs to regulate autophagy for asthma and COPD treatment.
Collapse
Affiliation(s)
- Giusy Daniela Albano
- Institute of Translational Pharmacology (IFT), National Research Council of Italy (CNR), Section of Palermo, Via Ugo La Malfa 153, 90146 Palermo, Italy; (A.M.M.); (R.G.); (M.P.)
| | | | | | | |
Collapse
|
10
|
Wei X, Wang Y, Lao Y, Weng J, Deng R, Li S, Lu J, Yang S, Liu X. Effects of honokiol protects against chronic kidney disease via BNIP3/NIX and FUNDC1-mediated mitophagy and AMPK pathways. Mol Biol Rep 2023; 50:6557-6568. [PMID: 37338733 DOI: 10.1007/s11033-023-08592-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023]
Abstract
BACKGROUND Chronic kidney disease (CKD) is a serious health threat worldwide. Defective mitophagy has been reported to induce mitochondrial dysfunction, which is closely associated with CKD pathogenesis. Honokiol (HKL) is a bioactive component of Magnolia officinalis that has multiple efficacies. Our study aimed to investigate the effect of HKL on a CKD rat model and explore the possible mechanisms of mitophagy mediated by Bcl-2 interacting protein 3 and BNIP3-like (NIX) (also known as the BNIP3/NIX pathway) and FUN14 domain-containing 1 (the FUNDC1 pathway) and the role of the AMP-activated protein kinase (AMPK) pathway. METHODS A CKD rat model was established by feeding the animals dietary adenine (0.75% w/w, 3 weeks). Simultaneously, the treatment group was given HKL (5 mg/kg/day, 4 weeks) by gavage. Renal function was assessed by measuring serum creatinine (Scr) and blood urea nitrogen (BUN) levels. Pathological changes were analyzed by periodic acid-Schiff (PAS) and Masson's trichrome staining. Protein expression was evaluated by Western blotting and immunohistochemistry. RESULTS HKL treatment ameliorated the decline in renal function and reduced tubular lesions and interstitial fibrosis in CKD rats. Accordingly, the renal fibrosis markers Col-IV and α-SMA were decreased by HKL. Moreover, HKL suppressed the upregulation of the proapoptotic proteins Bad and Bax and Cleaved caspase-3 expression in CKD rats. Furthermore, HKL suppressed BNIP3, NIX and FUNDC1 expression, leading to the reduction of excessive mitophagy in CKD rats. Additionally, AMPK was activated by adenine, and HKL reversed this change and significantly decreased the level of activated AMPK (phosphorylated AMPK, P-AMPK). CONCLUSION HKL exerted a renoprotective effect on CKD rats, which was possibly associated with BNIP3/NIX and FUNDC1-mediated mitophagy and the AMPK pathway.
Collapse
Affiliation(s)
- Xian Wei
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Yuzhi Wang
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, China
| | - Yunlan Lao
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, China
| | - Jiali Weng
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, China
| | - Ruyu Deng
- Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, 518000, Guangdong, China
| | - Shunmin Li
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Jiandong Lu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China
| | - Shudong Yang
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China.
| | - Xinhui Liu
- Department of Nephrology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518000, China.
| |
Collapse
|
11
|
Tulen CBM, Opperhuizen A, van Schooten FJ, Remels AHV. Disruption of the Molecular Regulation of Mitochondrial Metabolism in Airway and Lung Epithelial Cells by Cigarette Smoke: Are Aldehydes the Culprit? Cells 2023; 12:299. [PMID: 36672235 PMCID: PMC9857032 DOI: 10.3390/cells12020299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/23/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a devastating lung disease for which cigarette smoking is the main risk factor. Acetaldehyde, acrolein, and formaldehyde are short-chain aldehydes known to be formed during pyrolysis and combustion of tobacco and have been linked to respiratory toxicity. Mitochondrial dysfunction is suggested to be mechanistically and causally involved in the pathogenesis of smoking-associated lung diseases such as COPD. Cigarette smoke (CS) has been shown to impair the molecular regulation of mitochondrial metabolism and content in epithelial cells of the airways and lungs. Although it is unknown which specific chemicals present in CS are responsible for this, it has been suggested that aldehydes may be involved. Therefore, it has been proposed by the World Health Organization to regulate aldehydes in commercially-available cigarettes. In this review, we comprehensively describe and discuss the impact of acetaldehyde, acrolein, and formaldehyde on mitochondrial function and content and the molecular pathways controlling this (biogenesis versus mitophagy) in epithelial cells of the airways and lungs. In addition, potential therapeutic applications targeting (aldehyde-induced) mitochondrial dysfunction, as well as regulatory implications, and the necessary required future studies to provide scientific support for this regulation, have been covered in this review.
Collapse
Affiliation(s)
- Christy B. M. Tulen
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Antoon Opperhuizen
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Office of Risk Assessment and Research, Netherlands Food and Consumer Product Safety Authority, P.O. Box 43006, 3540 AA Utrecht, The Netherlands
| | - Frederik-Jan van Schooten
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Alexander H. V. Remels
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center+, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
12
|
Yang HH, Jiang HL, Tao JH, Zhang CY, Xiong JB, Yang JT, Liu YB, Zhong WJ, Guan XX, Duan JX, Zhang YF, Liu SK, Jiang JX, Zhou Y, Guan CX. Mitochondrial citrate accumulation drives alveolar epithelial cell necroptosis in lipopolysaccharide-induced acute lung injury. Exp Mol Med 2022; 54:2077-2091. [PMID: 36443565 PMCID: PMC9722936 DOI: 10.1038/s12276-022-00889-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/29/2022] Open
Abstract
Necroptosis is the major cause of death in alveolar epithelial cells (AECs) during acute lung injury (ALI). Here, we report a previously unrecognized mechanism for necroptosis. We found an accumulation of mitochondrial citrate (citratemt) in lipopolysaccharide (LPS)-treated AECs because of the downregulation of Idh3α and citrate carrier (CIC, also known as Slc25a1). shRNA- or inhibitor-mediated inhibition of Idh3α and Slc25a1 induced citratemt accumulation and necroptosis in vitro. Mice with AEC-specific Idh3α and Slc25a1 deficiency exhibited exacerbated lung injury and AEC necroptosis. Interestingly, the overexpression of Idh3α and Slc25a1 decreased citratemt levels and rescued AECs from necroptosis. Mechanistically, citratemt accumulation induced mitochondrial fission and excessive mitophagy in AECs. Furthermore, citratemt directly interacted with FUN14 domain-containing protein 1 (FUNDC1) and promoted the interaction of FUNDC1 with dynamin-related protein 1 (DRP1), leading to excessive mitophagy-mediated necroptosis and thereby initiating and promoting ALI. Importantly, necroptosis induced by citratemt accumulation was inhibited in FUNDC1-knockout AECs. We show that citratemt accumulation is a novel target for protection against ALI involving necroptosis.
Collapse
Affiliation(s)
- Hui-Hui Yang
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Hui-Ling Jiang
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Jia-Hao Tao
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Chen-Yu Zhang
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Jian-Bing Xiong
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Jin-Tong Yang
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Yu-Biao Liu
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Wen-Jing Zhong
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Xin-Xin Guan
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Jia-Xi Duan
- grid.216417.70000 0001 0379 7164Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Yan-Feng Zhang
- grid.216417.70000 0001 0379 7164Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Shao-Kun Liu
- grid.216417.70000 0001 0379 7164Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan China
| | - Jian-Xin Jiang
- grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns, and Combined Injury, Department of Trauma Medical Center, Daping Hospital, Army Medical University, Chongqing, China
| | - Yong Zhou
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| | - Cha-Xiang Guan
- grid.216417.70000 0001 0379 7164Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan China
| |
Collapse
|
13
|
Zhao X, Zhang Q, Zheng R. The interplay between oxidative stress and autophagy in chronic obstructive pulmonary disease. Front Physiol 2022; 13:1004275. [PMID: 36225291 PMCID: PMC9548529 DOI: 10.3389/fphys.2022.1004275] [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: 07/27/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Autophagy is a highly conserved process that is indispensable for cell survival, embryonic development, and tissue homeostasis. Activation of autophagy protects cells against oxidative stress and is a major adaptive response to injury. When autophagy is dysregulated by factors such as smoking, environmental insults and aging, it can lead to enhanced formation of aggressors and production of reactive oxygen species (ROS), resulting in oxidative stress and oxidative damage to cells. ROS activates autophagy, which in turn promotes cell adaptation and reduces oxidative damage by degrading and circulating damaged macromolecules and dysfunctional cell organelles. The cellular response triggered by oxidative stress includes changes in signaling pathways that ultimately regulate autophagy. Chronic obstructive pulmonary disease (COPD) is the most common lung disease among the elderly worldwide, with a high mortality rate. As an induced response to oxidative stress, autophagy plays an important role in the pathogenesis of COPD. This review discusses the regulation of oxidative stress and autophagy in COPD, and aims to provide new avenues for future research on target-specific treatments for COPD.
Collapse
Affiliation(s)
| | | | - Rui Zheng
- *Correspondence: Qiang Zhang, ; Rui Zheng,
| |
Collapse
|
14
|
Carinci M, Palumbo L, Pellielo G, Agyapong ED, Morciano G, Patergnani S, Giorgi C, Pinton P, Rimessi A. The Multifaceted Roles of Autophagy in Infectious, Obstructive, and Malignant Airway Diseases. Biomedicines 2022; 10:biomedicines10081944. [PMID: 36009490 PMCID: PMC9405571 DOI: 10.3390/biomedicines10081944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Autophagy is a highly conserved dynamic process by which cells deliver their contents to lysosomes for degradation, thus ensuring cell homeostasis. In response to environmental stress, the induction of autophagy is crucial for cell survival. The dysregulation of this degradative process has been implicated in a wide range of pathologies, including lung diseases, representing a relevant potential target with significant clinical outcomes. During lung disease progression and infections, autophagy may exert both protective and harmful effects on cells. In this review, we will explore the implications of autophagy and its selective forms in several lung infections, such as SARS-CoV-2, Respiratory Syncytial Virus (RSV) and Mycobacterium tuberculosis (Mtb) infections, and different lung diseases such as Cystic Fibrosis (CF), Chronic Obstructive Pulmonary Disease (COPD), and Malignant Mesothelioma (MM).
Collapse
Affiliation(s)
- Marianna Carinci
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Laura Palumbo
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Giulia Pellielo
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Esther Densu Agyapong
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Giampaolo Morciano
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Simone Patergnani
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Via Fossato di Mortara, 70, 44121 Ferrara, Italy
| | - Alessandro Rimessi
- Laboratory for Technologies of Advanced Therapies, Section of Experimental Medicine, Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center of Research for Innovative Therapies in Cystic Fibrosis, University of Ferrara, Via Fossato di Mortara, 70, 44121 Ferrara, Italy
- Correspondence:
| |
Collapse
|
15
|
Wang Z, Song Y, Jiang J, Piao Y, Li L, Bai Q, Xu C, Liu H, Li L, Piao H, Yan G. MicroRNA-182-5p Attenuates Asthmatic Airway Inflammation by Targeting NOX4. Front Immunol 2022; 13:853848. [PMID: 35711428 PMCID: PMC9192947 DOI: 10.3389/fimmu.2022.853848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Bronchial asthma is characterized by chronic airway inflammation, airway hyperresponsiveness, and airway remodeling. MicroRNA (miRNA) has recently been implicated in the pathogenesis of asthma. However, the mechanisms of different miRNAs in asthma are complicated, and the mechanism of miRNA-182-5p in asthma is still unclear. Here, we aim to explore the mechanism of miRNA182-5p in asthma-related airway inflammation. Ovalbumin (OVA)-induced asthma model was established. MiRNA Microarray Analysis was performed to analyze the differentially expressed miRNAs in the asthma model. We found that the expression of miRNA-182-5p was significantly decreased in OVA-induced asthma. In vitro, IL-13 stimulation of BEAS-2B cells resulted in a significant up-regulation of NOX4 (nicotinamide adenine dinucleotide phosphate oxidase 4), accompanied by mitochondrial damage-induced apoptosis, NLRP3 (NOD-like receptor family pyrin domain-containing 3)/IL-1β activation, and reduced miRNA-182-5p. In contrast, overexpression of miRNA-182-5p significantly inhibited epithelial cell apoptosis and NLRP3/IL-1β activation. In addition, we found that miRNA-182-5p could bind to the 3’ untranscripted region of NOX4 mRNA and inhibit epithelial cell inflammation by reducing oxidative stress and mitochondrial damage. In vivo, miRNA-182-5p agomir treatment significantly reduced the percentage of eosinophils in bronchoalveolar lavage fluid, and down-regulated Th2 inflammatory factors, including IL-4, IL-5, and OVA induced IL-13. Meanwhile, miRNA-182-5p agomir reduced the peribronchial inflammatory cell infiltration, goblet cell proliferation and collagen deposition. In summary, targeting miRNA-182-5p may provide a new strategy for the treatment of asthma.
Collapse
Affiliation(s)
- Zhiguang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yilan Song
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Jingzhi Jiang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Yihua Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Intensive Care Unit, Affiliated Hospital of Yanbian University, Yanji, China
| | - Li Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Qiaoyun Bai
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Chang Xu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Hanye Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Hongmei Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, China
- *Correspondence: Hongmei Piao, ; Guanghai Yan, ;
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China
- Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
- *Correspondence: Hongmei Piao, ; Guanghai Yan, ;
| |
Collapse
|
16
|
MTMR14 Alleviates Chronic Obstructive Pulmonary Disease as a Regulator in Inflammation and Emphysema. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9300269. [PMID: 35035670 PMCID: PMC8759842 DOI: 10.1155/2022/9300269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023]
Abstract
Extensive inflammation and apoptosis in structural cells of the lung are responsible for the progression and pathogenesis of chronic obstructive pulmonary disease (COPD). Myotubularin-related protein 14 (MTMR14) has been shown to participate in various biological processes, including apoptosis, inflammation, and autophagy. Nonetheless, the role of MTMR14 in COPD remains elusive. In the present study, we explored the expression of MTMR14 in human lung tissues and investigated the effects of overexpressed MTMR14 on in vitro and in vivo COPD models. Moreover, one of the possible mechanisms of MTMR14 alleviating COPD was explored based on mitochondrial function and mitophagy homeostasis. The results showed that MTMR14 expression was reduced in COPD patients' lungs in comparison to control subjects. MTMR14 overexpression inhibited cigarette smoke extract-induced inflammation and apoptosis and improved mitochondrial function and mitophagy in vitro. Further verification was carried out in COPD model mice. MTMR14 overexpression inhibited lung inflammation and reduced levels of IL-6 and KC in bronchoalveolar lavage fluid, as well as prevented emphysema and a decline in lung function. Furthermore, MTMR14 overexpression improved mitochondrial function and mitophagy to a certain extent. Collectively, our data support the hypothesis that MTMR14 participates in the pathogenesis of COPD. Improving mitochondrial function and mitophagy homeostasis may be one of the mechanisms by which MTMR14 alleviates COPD and may potentially be a novel therapeutic target for COPD.
Collapse
|
17
|
Tulen CBM, Wang Y, Beentjes D, Jessen PJJ, Ninaber DK, Reynaert NL, van Schooten FJ, Opperhuizen A, Hiemstra PS, Remels AHV. Dysregulated mitochondrial metabolism upon cigarette smoke exposure in various human bronchial epithelial cell models. Dis Model Mech 2022; 15:dmm049247. [PMID: 35344036 PMCID: PMC8990921 DOI: 10.1242/dmm.049247] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/29/2021] [Indexed: 01/13/2023] Open
Abstract
Exposure to cigarette smoke (CS) is the primary risk factor for developing chronic obstructive pulmonary disease. The impact of CS exposure on the molecular mechanisms involved in mitochondrial quality control in airway epithelial cells is incompletely understood. Undifferentiated or differentiated primary bronchial epithelial cells were acutely/chronically exposed to whole CS (WCS) or CS extract (CSE) in submerged or air-liquid interface conditions. Abundance of key regulators controlling mitochondrial biogenesis, mitophagy and mitochondrial dynamics was assessed. Acute exposure to WCS or CSE increased the abundance of components of autophagy and receptor-mediated mitophagy in all models. Although mitochondrial content and dynamics appeared to be unaltered in response to CS, changes in both the molecular control of mitochondrial biogenesis and a shift toward an increased glycolytic metabolism were observed in particular in differentiated cultures. These alterations persisted, at least in part, after chronic exposure to WCS during differentiation and upon subsequent discontinuation of WCS exposure. In conclusion, smoke exposure alters the regulation of mitochondrial metabolism in airway epithelial cells, but observed alterations may differ between various culture models used. This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Christy B. M. Tulen
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Ying Wang
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Daan Beentjes
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Phyllis J. J. Jessen
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Dennis K. Ninaber
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Niki L. Reynaert
- Department of Respiratory Medicine, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Primary Lung Culture Facility, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Frederik-Jan van Schooten
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Antoon Opperhuizen
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Office of Risk Assessment and Research, Netherlands Food and Consumer Product Safety Authority, PO Box 8433, 3503 RK Utrecht, The Netherlands
| | - Pieter S. Hiemstra
- Department of Pulmonology, Leiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Alexander H. V. Remels
- Department of Pharmacology and Toxicology, School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
18
|
Dong D, Wu J, Sheng L, Gong X, Zhang Z, Yu C. FUNDC1 induces apoptosis and autophagy under oxidative stress via PI3K/Akt/mTOR pathway in cataract lens cells. Curr Eye Res 2022; 47:547-554. [PMID: 35179404 DOI: 10.1080/02713683.2021.2021586] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVE This purpose of the study is to explore the mRNA and protein expression of FUNDC1 in cataract cells and tissues, clarify the function and mechanism of FUNDC1 in cataract cells under oxidative stress. METHODS We used bioinformatic analysis to screen DEGs in cataract from GSE153933. The expression of FUNDC1 in cataract specimens and cells was measured by RT-qPCR and western blotting. MethPrimer was used to predict CpG island of FUNDC1 promoter. The methylation of FUNDC1 in cataract specimens and cells was determined by MSP assay. Flow cytometry assay was used to measure cell apoptosis in FUNDC1-knockdown and -overexpression SRA01/04 cells. The expression of LC3 was analyzed by immunofluorescence assay. The expression of apoptosis related proteins, autophagy and PI3K/Akt/mTOR related proteins was determined by western blotting. RESULTS The results of bioinformatic analysis revealed that FUNDC1 was upregulation in cataract. FUNDC1 was further high expression in SRA01/04 cells with H2O2 treatment whereas hypomethylation of FUNDC1 in cataract lens cells under oxidative stress. Knockdown of FUNDC1 decreased cell apoptosis and autophagy compared with negative control of SRA01/04 cells. While overexpression of FUNDC1 elevated cell apoptosis and autophagy compared to empty vector group in SRA01/04 cells. Mechanically, FUNDC1 reduced the phosphorylation of PI3K/Akt/mTOR pathway under oxidative stress in SRA01/04 cells. CONCLUSION Our study suggested that FUNDC1 deficiency restrains cell apoptosis and autophagy through inhibiting PI3K/Akt/mTOR signal pathway.
Collapse
Affiliation(s)
- Duo Dong
- Department of Ophthalmology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang Province, 161000, China
| | - Jing Wu
- Department of Ophthalmology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang Province, 161000, China
| | - Lijie Sheng
- Department of Ophthalmology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang Province, 161000, China
| | - Xuewu Gong
- Department of Ophthalmology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang Province, 161000, China
| | - Zhichang Zhang
- Department of Ophthalmology, The Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, Heilongjiang Province, 161000, China
| | - Caihan Yu
- Department of Ophthalmology, Xianning Central Hospital, The first Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei Province, 437100, China
| |
Collapse
|
19
|
Wang L, Jiang W, Wang J, Xie Y, Wang W. Puerarin inhibits FUNDC1-mediated mitochondrial autophagy and CSE-induced apoptosis of human bronchial epithelial cells by activating the PI3K/AKT/mTOR signaling pathway. Aging (Albany NY) 2022; 14:1253-1264. [PMID: 35134750 PMCID: PMC8876910 DOI: 10.18632/aging.203317] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/27/2021] [Indexed: 12/30/2022]
Abstract
Increasing evidence suggests that the pathogenesis of chronic obstructive pulmonary disease (COPD) is associated with FUN14 domain protein 1 (FUNDC1)-mediated mitophagy. Recently, studies have reported that puerarin has protective effects against excessive oxidative damage in cells. Therefore, we hypothesized that puerarin may be involved in COPD progression via regulating FUNDC1 mediated mitophagy. We found that the viability of cigarette smoke extract (CSE)-stimulated human bronchial epithelial cells (HBECs) was enhanced and apoptosis was reduced after treatment with different concentrations of puerarin. Puerarin reversed mitochondrial membrane potential (MMP) levels and ATP content, and decreased reactive oxygen species (ROS) content in CSE stimulated HBECs. Moreover, puerarin significantly inhibited apoptosis related proteins, as well as the expression of mitophagy related proteins. After inhibition of FUNDC1 phosphorylation by protein phosphatase inhibitor (PH0321), puerarin restored MMP level, decreased ROS content, promoted ATP synthesis, and downregulated autophagy related protein expression in HBECs. In addition, mitochondrial division inhibitor (Mdivi) inhibited the expression of autophagy related proteins and reduced apoptosis after blocking cell autophagy, which was the same as the inhibition of puerarin. Finally, puerarin activated the PI3K/Akt/mTOR signaling pathway to participate in COPD progression by up regulating the phosphorylation levels of PI3K, Akt and mTOR.
Collapse
Affiliation(s)
- Li Wang
- Department of Respiratory Medicine, Yan’an University Affiliated Hospital, Yan’an 716000, China
| | - Weizhou Jiang
- Department of Pulmonology, Weifang Traditional Chinese Hospital, Weifang 261041, China
| | - Jing Wang
- Endoscopy Room, Tai’an Maternal and Child Health Hospital, Tai’an 271000, China
| | - Yuanyuan Xie
- Department of Geriatrics, Yan’an University Affiliated Hospital, Yan’an 716000, China
| | - Weisi Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, China
| |
Collapse
|
20
|
Jiang H, Jiang Y, Xu Y, Yuan D, Li Y. Bronchial epithelial SIRT1 deficiency exacerbates cigarette smoke induced emphysema in mice through the FOXO3/PINK1 pathway. Exp Lung Res 2022:1-16. [PMID: 35132913 DOI: 10.1080/01902148.2022.2037169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/17/2022] [Accepted: 01/29/2022] [Indexed: 11/04/2022]
Abstract
Purpose: Cellular senescence and mitochondrial fragmentation are thought to be crucial components of the cigarette smoke(CS)-induced responses that contribute to the chronic obstructive pulmonary disease (COPD) development as a result of accelerated premature aging of the lung. Although there have been a few reports on the role of sirtuin 1(SIRT1) in mitochondrial homeostasis, senescence and inflammation, whether SIRT1/FOXO3/PINK1 signaling mediated mitophagy ameliorates cellular senescence in COPD is still unclear. This study aimed to ascertain whether SIRT1 regulates cellular senescence via FOXO3/PINK1-mediated mitophagy in COPD. Methods: To investigate the effect of CS exposure and SIRT1 deficiency on mitophagy and senescence in the lung, a SIRT1 knockout(KO) mouse model was used. Airway resistance, cellular senescence mitochondrial injury, mitophagy, cellular architecture and protein expression levels in lung tissues, from SIRT1 KO and wild-type(WT) COPD model mice exposed to CS for 6 months were examined by western blotting, histochemistry, immunofluorescence and transmission electron microscopy(TEM). Results: In CS exposed mice, SIRT1 deficiency exacerbated airway resistance and cellular senescence, increased FOXO3 acetylation and decreased PINK1 protein levels and attenuated mitophagy. Mechanistically, the damaging effect of SIRT1 deficiency on lung tissue was attributed to increased FOXO3 acetylation and decreased PINK1 levels, and attenuated mitophagy. In vitro, mitochondrial damage and cellular sensitivity in response to CS exposure were more severe in control cells than in cells treated with aSIRT1 activator. SIRT1 activation SIRT1 activation decreased FOXO3 acetylation and increased the protein levels of PINK1 and enhanced mitophagy. Conclusion: These results demonstrated that the detrimental effects of SIRT1 deficiency on cell senescence associated with insufficient mitophagy, and involved the FOXO3/PINK1 signaling pathway.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Clinical Medicine, Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Internal Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yaona Jiang
- Department of Internal Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
- Graduate Department, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yuanri Xu
- Department of Internal Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
- Graduate Department, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Dong Yuan
- Department of Internal Medicine, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yaqing Li
- Department of Internal Medicine, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
Liu H, Zang C, Yuan F, Ju C, Shang M, Ning J, Yang Y, Ma J, Li G, Bao X, Zhang D. The role of FUNDC1 in mitophagy, mitochondrial dynamics and human diseases. Biochem Pharmacol 2021; 197:114891. [PMID: 34968482 DOI: 10.1016/j.bcp.2021.114891] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/01/2021] [Accepted: 12/18/2021] [Indexed: 12/22/2022]
Abstract
Mitochondria are the principal sites of energy metabolism and provide most of the energy needed for normal cellular function. They are dynamic organelles that constantly undergo fission, fusion and mitophagy to maintain their homeostasis and function. However, dysregulated mitochondrial dynamics and mitophagy leads to reduced ATP generation and mutation of their DNA, which ultimately leads to cell death. Increasing evidence has shown that the FUN14 domain-containing protein 1 (FUNDC1), a novel mitophagy receptor, participates in the process of mitochondrial dynamics and mitophagy and plays a critical role in various human diseases. Herein, we review the role of FUNDC1 in mitophagy and mitochondrial dynamics, thus providing a better understanding of the relationship between the two processes. Moreover, we summarize the treatments targeting FUNDC1, and suggest that FUNDC1 may represent a promising therapeutic target for the treatment of several human diseases such as cardiovascular diseases, metabolic syndrome, cancer and chronic obstructive pulmonary disease (COPD).
Collapse
Affiliation(s)
- Hui Liu
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Caixia Zang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Fangyu Yuan
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Cheng Ju
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Meiyu Shang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Jingwen Ning
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Yang Yang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Jingwei Ma
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Gen Li
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China
| | - Xiuqi Bao
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China.
| | - Dan Zhang
- State Key Laboratory of Bioactive Substrate and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing 100050, China.
| |
Collapse
|
22
|
Chen G, Chen L, Huang Y, Zhu X, Yu Y. Increased FUN14 domain containing 1 (FUNDC1) ubiquitination level inhibits mitophagy and alleviates the injury in hypoxia-induced trophoblast cells. Bioengineered 2021; 13:3620-3633. [PMID: 34699308 PMCID: PMC8974051 DOI: 10.1080/21655979.2021.1997132] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Preeclampsia (PE) is a pregnancy disorder characterized by excessive trophoblast cell death. This study aims to explore the exact mechanism of the ubiquitination level of FUN14 domain containing 1 (FUNDC1) in mitophagy and injury in hypoxic trophoblast cells. In this study, HTR-8/SVneo trophoblast cells were cultured under normoxic and hypoxic conditions and PE mouse model was established. We found low ubiquitination level of FUNDC1 in hypoxic trophoblast cells and placenta of pregnant women with PE. Proteasome inhibitor MG-132 and protease activator MF-094 were added into HTR-8/SVneo trophoblast cells. Proteasome inhibitor MG-132 decreased FUNDC1 ubiquitination level while protease activator MF-094 increased FUNDC1 ubiquitination level. Inhibition of FUNDC1 ubiquitination promoted mitophagy and mitochondrial membrane potential (Δψm) in normoxic trophoblast cells, increased levels of reactive oxygen species (ROS) and malondialdehyde (MDA) and decreased levels of glutathione (GSH) and superoxide dismutase (SOD). In addition, FUNDC1 ubiquitination alleviated cell injury in PE mice in vivo. In conclusion, increased FUNDC1 ubiquitination level inhibited mitophagy and Δψm changes in hypoxic trophoblast cells, and thus alleviated oxidative injury.
Collapse
Affiliation(s)
- GuoQing Chen
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong 518028; China
| | - Lu Chen
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong 518028; China
| | - Yan Huang
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong 518028; China
| | - XiongShan Zhu
- Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong 518028; China
| | - YuanLan Yu
- Department of Emergency, Shenzhen Children's Hospital, Shenzhen, Guangdong 518026, China
| |
Collapse
|
23
|
Mitochondrial Dysfunction in Chronic Respiratory Diseases: Implications for the Pathogenesis and Potential Therapeutics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5188306. [PMID: 34354793 PMCID: PMC8331273 DOI: 10.1155/2021/5188306] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/30/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023]
Abstract
Mitochondria are indispensable for energy metabolism and cell signaling. Mitochondrial homeostasis is sustained with stabilization of mitochondrial membrane potential, balance of mitochondrial calcium, integrity of mitochondrial DNA, and timely clearance of damaged mitochondria via mitophagy. Mitochondrial dysfunction is featured by increased generation of mitochondrial reactive oxygen species, reduced mitochondrial membrane potential, mitochondrial calcium imbalance, mitochondrial DNA damage, and abnormal mitophagy. Accumulating evidence indicates that mitochondrial dysregulation causes oxidative stress, inflammasome activation, apoptosis, senescence, and metabolic reprogramming. All these cellular processes participate in the pathogenesis and progression of chronic respiratory diseases, including chronic obstructive pulmonary disease, pulmonary fibrosis, and asthma. In this review, we provide a comprehensive and updated overview of the impact of mitochondrial dysfunction on cellular processes involved in the development of these respiratory diseases. This not only implicates mechanisms of mitochondrial dysfunction for the pathogenesis of chronic lung diseases but also provides potential therapeutic approaches for these diseases by targeting dysfunctional mitochondria.
Collapse
|
24
|
Zhu H, Toan S, Mui D, Zhou H. Mitochondrial quality surveillance as a therapeutic target in myocardial infarction. Acta Physiol (Oxf) 2021; 231:e13590. [PMID: 33270362 DOI: 10.1111/apha.13590] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/06/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022]
Abstract
Myocardial infarction (MI) is a leading cause of morbidity and mortality worldwide. As mitochondrial dysfunction critically contributes to the pathogenesis of MI, intensive research is focused on the development of therapeutic strategies targeting mitochondrial homeostasis. Mitochondria possess a quality control system which maintains and restores their structure and function by regulating mitochondrial fission, fusion, biogenesis, degradation and death. In response to slight damage such as transient hypoxia or mild oxidative stress, mitochondrial metabolism shifts from oxidative phosphorylation to glycolysis, in order to reduce oxygen consumption and maintain ATP output. Mitochondrial dynamics are also activated to modify mitochondrial shape and structure, in order to meet cardiomyocyte energy requirements through augmenting or reducing mitochondrial mass. When damaged mitochondria cannot be repaired, poorly structured mitochondria will be degraded through mitophagy, a process which is often accompanied by mitochondrial biogenesis. Once the insult is severe enough to induce lethal damage in the mitochondria and the cell, mitochondrial death pathway activation is an inevitable consequence, and the cardiomyocyte apoptosis or necrosis program will be initiated to remove damaged cells. Mitochondrial quality surveillance is a hierarchical system preserving mitochondrial function and defending cardiomyocytes against stress. A failure of this system has been regarded as one of the potential pathologies underlying MI. In this review, we discuss the recent findings focusing on the role of mitochondrial quality surveillance in MI, and highlight the available therapeutic approaches targeting mitochondrial quality surveillance during MI.
Collapse
Affiliation(s)
- Hang Zhu
- Department of Cardiology Chinese PLA General HospitalMedical School of Chinese PLA Beijing China
| | - Sam Toan
- Department of Chemical Engineering University of Minnesota‐Duluth Duluth MN USA
| | - David Mui
- Perelman School of Medicine University of Pennsylvania Philadelphia PA USA
| | - Hao Zhou
- Department of Cardiology Chinese PLA General HospitalMedical School of Chinese PLA Beijing China
| |
Collapse
|
25
|
Sun S, Shen Y, Wang J, Li J, Cao J, Zhang J. Identification and Validation of Autophagy-Related Genes in Chronic Obstructive Pulmonary Disease. Int J Chron Obstruct Pulmon Dis 2021; 16:67-78. [PMID: 33469280 PMCID: PMC7811454 DOI: 10.2147/copd.s288428] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/30/2020] [Indexed: 12/19/2022] Open
Abstract
Purpose Autophagy plays essential roles in the development of COPD. We aim to identify and validate the potential autophagy-related genes of COPD through bioinformatics analysis and experiment validation. Methods The mRNA expression profile dataset GSE38974 was obtained from GEO database. The potential differentially expressed autophagy-related genes of COPD were screened by R software. Then, protein–protein interactions (PPI), correlation analysis, gene-ontology (GO) enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were applied for the differentially expressed autophagy-related genes. Finally, RNA expression of top five differentially expressed autophagy-related genes was validated in blood samples from COPD patients and healthy controls by qRT-PCR. Results A total of 40 differentially expressed autophagy-related genes (14 up-regulated genes and 26 down-regulated genes) were identified between 23 COPD patients and 9 healthy controls. The PPI results demonstrated that these autophagy-related genes interacted with each other. The GO and KEGG enrichment analysis of differentially expressed autophagy-related genes indicated several enriched terms related to autophagy and mitophagy. The results of qRT-PCR showed that the expression levels of HIF1A, CDKN1A, BAG3, ERBB2 and ATG16L1 in COPD patients and healthy controls were consistent with the bioinformatics analysis results from mRNA microarray. Conclusion We identified 40 potential autophagy-related genes of COPD through bioinformatics analysis. HIF1A, CDKN1A, BAG3, ERBB2 and ATG16L1 may affect the development of COPD by regulating autophagy. These results may expand our understanding of COPD and might be useful in the treatment of COPD.
Collapse
Affiliation(s)
- Shulei Sun
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China
| | - Yuehao Shen
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China
| | - Jie Wang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China
| | - Jinna Li
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China
| | - Jie Cao
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China
| | - Jing Zhang
- Department of Respiratory and Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, People's Republic of China
| |
Collapse
|
26
|
Racanelli AC, Choi AMK, Choi ME. Autophagy in chronic lung disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 172:135-156. [PMID: 32620240 DOI: 10.1016/bs.pmbts.2020.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The development of chronic lung disease occurs as a consequence of multiple cellular events that involve an initial insult which often leads to the development of chronic inflammation, and the dysregulation of cellular proliferation and cell death mechanisms. Multiple cell types in the lung are key to the respiratory and protective/barrier functions necessary to manage the chronic exposures to environmental, mechanical, and oxidative stressors. Autophagy is essential to lung development and homeostasis, as well as the prevention and development of disease. The cellular process involves the collection and removal of unwanted organelles and proteins through lysosomal degradation. In recent years, investigations have addressed the roles of autophagy and selective autophagy in numerous chronic lung diseases. Here, we highlight recent advances on the role of autophagy in the pathogenesis of asthma, chronic obstructive pulmonary disease and emphysema, pulmonary arterial hypertension, and idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Alexandra C Racanelli
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States; NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States; NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States
| | - Mary E Choi
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, NY, United States; Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|