1
|
Chen Y, Liu J, Qin H, Qin S, Huang X, Wei C, Hu X. Deciphering regulatory patterns in a mouse model of hyperoxia-induced acute lung injury. PeerJ 2024; 12:e18069. [PMID: 39346085 PMCID: PMC11439394 DOI: 10.7717/peerj.18069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 08/19/2024] [Indexed: 10/01/2024] Open
Abstract
Background Oxygen therapy plays a pivotal role in treating critically ill patients in the intensive care unit (ICU). However, excessive oxygen concentrations can precipitate hyperoxia, leading to damage in multiple organs, with a notable effect on the lungs. Hyperoxia condition may lead to hyperoxia-induced acute lung injury (HALI), deemed as a milder form of acute respiratory distress syndrome (ARDS). Given its clinical importance and practical implications, there is a compelling need to investigate the underlying pathogenesis and comprehensively understand the regulatory mechanisms implicated in the development of HALI. Results In this study, we conducted a mouse model with HALI and performed regulatory mechanism analysis using RNA-seq on both HALI and control group. Comprehensive analysis revealed 727 genes of significant differential expression, including 248 long non-coding RNAs (lncRNAs). Also, alternative splicing events were identified from sequencing results. Notably, we observed up-regulation or abnormal alternative splicing of genes associated with immune response and ferroptosis under hyperoxia conditions. Utilizing weighted gene co-expression network analysis (WGCNA), we ascertained that genes involved in immune response formed a distinct cluster, showcasing an up-regulated pattern in hyperoxia, consistent with previous studies. Furthermore, a competing endogenous RNA (ceRNA) network was constructed, including 78 differentially expressed mRNAs and six differentially expressed lncRNAs, including H19. These findings uncover the intricate interplay of multiple transcriptional regulatory mechanisms specifically tailored to the pulmonary defense against HALI, substantiating the importance of these non-coding RNAs in this disease context. Conclusions Our results provide new insights into the potential mechanisms and underlying pathogenesis in the development of HALI at the post-transcriptional level. The findings of this study reveal potential regulatory interactions and biological roles of specific lncRNAs and genes, such as H19 and Sox9, encompassing driven gene expression patterns, alternative splicing events, and lncRNA-miRNA-mRNA ceRNA networks. These findings may pave the way for advancing therapeutic strategies and reducing the risk associated with oxygen treatment for patients.
Collapse
Affiliation(s)
- Yundi Chen
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinwen Liu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Nutrition, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Qin
- Department of Respiratory and Critical Care Medicine, Kweichow Moutai Hospital, Zunyi, Guizhou, China
| | - Song Qin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Xinyang Huang
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunyan Wei
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaolin Hu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Narala VR, Narala SR, Aiya Subramani P, Panati K, Kolliputi N. Role of mitochondria in inflammatory lung diseases. Front Pharmacol 2024; 15:1433961. [PMID: 39228517 PMCID: PMC11368744 DOI: 10.3389/fphar.2024.1433961] [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: 05/16/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
Mitochondria play a significant and varied role in inflammatory lung disorders. Mitochondria, known as the powerhouse of the cell because of their role in producing energy, are now recognized as crucial regulators of inflammation and immunological responses. Asthma, chronic obstructive pulmonary disease, and acute respiratory distress syndrome are characterized by complex interactions between immune cells, inflammatory substances, and tissue damage. Dysfunctional mitochondria can increase the generation of reactive oxygen species (ROS), triggering inflammatory pathways. Moreover, mitochondrial failure impacts cellular signaling, which in turn affects the expression of molecules that promote inflammation. In addition, mitochondria have a crucial role in controlling the behavior of immune cells, such as their activation and differentiation, which is essential in the development of inflammatory lung diseases. Their dynamic behavior, encompassing fusion, fission, and mitophagy, also impacts cellular responses to inflammation and oxidative stress. Gaining a comprehensive understanding of the intricate correlation between mitochondria and lung inflammation is essential in order to develop accurate treatment strategies. Targeting ROS generation, dynamics, and mitochondrial function may offer novel approaches to treating inflammatory lung diseases while minimizing tissue damage. Additional investigation into the precise contributions of mitochondria to lung inflammation will provide significant knowledge regarding disease mechanisms and potential therapeutic approaches. This review will focus on how mitochondria in the lung regulate these processes and their involvement in acute and chronic lung diseases.
Collapse
Affiliation(s)
| | | | | | - Kalpana Panati
- Department of Biotechnology, Government College for Men, Kadapa, India
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
3
|
Cattani-Cavalieri I, Trombetta-Lima M, Yan H, Manzano-Covarrubias AL, Baarsma HA, Oun A, van der Veen MM, Oosterhout E, Dolga AM, Ostrom RS, Valenca SS, Schmidt M. Diesel exhaust particles alter mitochondrial bioenergetics and cAMP producing capacity in human bronchial epithelial cells. FRONTIERS IN TOXICOLOGY 2024; 6:1412864. [PMID: 39118833 PMCID: PMC11306203 DOI: 10.3389/ftox.2024.1412864] [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: 04/09/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Introduction: Air pollution from diesel combustion is linked in part to the generation of diesel exhaust particles (DEP). DEP exposure induces various processes, including inflammation and oxidative stress, which ultimately contribute to a decline in lung function. Cyclic AMP (cAMP) signaling is critical for lung homeostasis. The impact of DEP on cAMP signaling is largely unknown. Methods: We exposed human bronchial epithelial (BEAS-2B) cells to DEP for 24-72 h and evaluated mitochondrial bioenergetics, markers of oxidative stress and inflammation and the components of cAMP signaling. Mitochondrial bioenergetics was measured at 72 h to capture the potential and accumulative effects of prolonged DEP exposure on mitochondrial function. Results: DEP profoundly altered mitochondrial morphology and network integrity, reduced both basal and ATP-linked respiration as well as the glycolytic capacity of mitochondria. DEP exposure increased gene expression of oxidative stress and inflammation markers such as interleukin-8 and interleukin-6. DEP significantly affected mRNA levels of exchange protein directly activated by cAMP-1 and -2 (Epac1, Epac2), appeared to increase Epac1 protein, but left phospho-PKA levels unhanged. DEP exposure increased A-kinase anchoring protein 1, β2-adrenoceptor and prostanoid E receptor subtype 4 mRNA levels. Interestingly, DEP decreased mRNA levels of adenylyl cyclase 9 and reduced cAMP levels stimulated by forskolin (AC activator), fenoterol (β2-AR agonist) or PGE2 (EPR agonist). Discussion: Our findings suggest that DEP induces mitochondrial dysfunction, a process accompanied by oxidative stress and inflammation, and broadly dampens cAMP signaling. These epithelial responses may contribute to lung dysfunction induced by air pollution exposure.
Collapse
Affiliation(s)
- Isabella Cattani-Cavalieri
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Marina Trombetta-Lima
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, Netherlands
| | - Hong Yan
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Ana L. Manzano-Covarrubias
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hoeke A. Baarsma
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Asmaa Oun
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | | | - Emily Oosterhout
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Amalia M. Dolga
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rennolds S. Ostrom
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Samuel Santos Valenca
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
4
|
Sun J, Shao Y, Pei L, Zhu Q, Yu X, Yao W. AKAP1 alleviates VSMC phenotypic modulation and neointima formation by inhibiting Drp1-dependent mitochondrial fission. Biomed Pharmacother 2024; 176:116858. [PMID: 38850669 DOI: 10.1016/j.biopha.2024.116858] [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: 12/18/2023] [Revised: 05/26/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
The roles and mechanisms of A-kinase anchoring protein 1 (AKAP1) in vascular smooth muscle cell (VSMC) phenotypic modulation and neointima formation are currently unknown. AKAP1 is a mitochondrial PKA-anchored protein and maintains mitochondrial homeostasis. This study aimed to investigate how AKAP1/PKA signaling plays a protective role in inhibiting VSMC phenotypic transformation and neointima formation by regulating mitochondrial fission. The results showed that both PDGF-BB treatment and balloon injury reduced the transcription, expression, and mitochondrial anchoring of AKAP1. In vitro, the overexpression of AKAP1 significantly inhibited PDGF-BB mediated VSMC proliferation and migration, whereas AKAP1 knockdown further aggravated VSMC phenotypic transformation. Additionally, in the balloon injury model in vivo, AKAP1 overexpression reduced neointima formation, the muscle fiber area ratio, and rat VSMC proliferation and migration. Furthermore, PDGF-BB and balloon injury inhibited Drp1 phosphorylation at Ser637 and promoted Drp1 activity and mitochondrial midzone fission; AKAP1 overexpression reversed these effects. AKAP1 overexpression also inhibited the distribution of mitochondria at the plasma membrane and the reduction of PKARIIβ expression induced by PDGF-BB, as evidenced by an increase in mitochondria-plasma membrane distance as well as PKARIIβ protein levels. Moreover, the PKA agonist promoted Drp1 phosphorylation (Ser637) and inhibited PDGF-BB-mediated mitochondrial fission, cell proliferation, and migration. The PKA antagonist reversed the increase in Drp1 phosphorylation (Ser637) and the decline in mitochondrial midzone fission and VSMC phenotypic transformation caused by AKAP1 overexpression. The results of this study reveal that AKAP1 protects VSMCs against phenotypic modulation by improving Drp1 phosphorylation at Ser637 through PKA and inhibiting mitochondrial fission, thereby preventing neointima formation.
Collapse
MESH Headings
- Animals
- Male
- Rats
- A Kinase Anchor Proteins/metabolism
- A Kinase Anchor Proteins/genetics
- Becaplermin/pharmacology
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Dynamins/metabolism
- Mitochondrial Dynamics/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Neointima/metabolism
- Neointima/pathology
- Phenotype
- Phosphorylation
- Rats, Sprague-Dawley
- Signal Transduction
Collapse
Affiliation(s)
- Jingwen Sun
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Yuting Shao
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Lele Pei
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Qingyu Zhu
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China
| | - Xiaoqiang Yu
- Department of Vascular Surgery, The First People's Hospital of Nantong, Nantong 226001, China
| | - Wenjuan Yao
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong 226001, China.
| |
Collapse
|
5
|
Jeong J, Lee Y, Han J, Kang E, Kim D, Kim KS, Kim EAR, Lee BS, Jung E. Mitochondrial DNA mutations in extremely preterm infants with bronchopulmonary dysplasia. Gene 2024; 910:148337. [PMID: 38432533 DOI: 10.1016/j.gene.2024.148337] [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: 10/01/2023] [Revised: 02/21/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
Bronchopulmonary dysplasia (BPD) is a serious chronic lung disease affecting extremely preterm infants. While mitochondrial dysfunction has been investigated in various medical conditions, limited research has explored mitochondrial DNA (mtDNA) gene mutations, specifically in BPD. This study aimed to evaluate mitochondrial mtDNA gene mutations in extremely preterm infants with BPD. In this prospective observational study, we enrolled a cohort of extremely preterm infants diagnosed with BPD. Clinical data were collected to provide comprehensive patient profiles. Peripheral blood mononuclear cells were isolated from whole-blood samples obtained within a defined timeframe. Subsequently, mtDNA extraction and sequencing using next-generation sequencing technology were performed to identify mtDNA gene mutations. Among the cohort of ten extremely preterm infants with BPD, mtDNA sequencing revealed the presence of mutations in seven patients, resulting in a total of twenty-one point mutations. Notably, many of these mutations were identified in loci associated with critical components of the respiratory chain complexes, vital for proper mitochondrial function and cellular energy production. This pilot study provides evidence of mtDNA point mutations in a subset of extremely preterm infants with BPD. These findings suggest a potential association between mitochondrial dysfunction and the pathogenesis of BPD. Further extensive investigations are warranted to unravel the mechanisms underlying mtDNA mutations in BPD.
Collapse
Affiliation(s)
- Jiyoon Jeong
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Yeonmi Lee
- Department of Convergence Medicine and Stem Cell Center, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea; Department of Biomedical Science, College of Life Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea.
| | - Jongsuk Han
- Department of Convergence Medicine and Stem Cell Center, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea; Department of Biomedical Science, College of Life Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea.
| | - Eunju Kang
- Department of Convergence Medicine and Stem Cell Center, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea; Department of Biomedical Science, College of Life Science, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, Republic of Korea.
| | - Deokhoon Kim
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Ki-Soo Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Ellen Ai-Rhan Kim
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Byong Sop Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| | - Euiseok Jung
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, 88, Olympic-ro 43-gil, Songpa-gu, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Wang X, Qin S, Ren Y, Feng B, Liu J, Yu K, Yu H, Liao Z, Mei H, Tan M. Gpnmb silencing protects against hyperoxia-induced acute lung injury by inhibition of mitochondrial-mediated apoptosis. Hum Exp Toxicol 2024; 43:9603271231222873. [PMID: 38166464 DOI: 10.1177/09603271231222873] [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] [Indexed: 01/04/2024]
Abstract
Background: Hyperoxia-induced acute lung injury (HALI) is a complication to ventilation in patients with respiratory failure, which can lead to acute inflammatory lung injury and chronic lung disease. The aim of this study was to integrate bioinformatics analysis to identify key genes associated with HALI and validate their role in H2O2-induced cell injury model.Methods: Integrated bioinformatics analysis was performed to screen vital genes involved in hyperoxia-induced lung injury (HLI). CCK-8 and flow cytometry assays were performed to assess cell viability and apoptosis. Western blotting was performed to assess protein expression.Results: In this study, glycoprotein non-metastatic melanoma protein B (Gpnmb) was identified as a key gene in HLI by integrated bioinformatics analysis of 4 Gene Expression Omnibus (GEO) datasets (GSE97804, GSE51039, GSE76301 and GSE87350). Knockdown of Gpnmb increased cell viability and decreased apoptosis in H2O2-treated MLE-12 cells, suggesting that Gpnmb was a proapoptotic gene during HALI. Western blotting results showed that knockdown of Gpnmb reduced the expression of Bcl-2 associated X (BAX) and cleaved-caspase 3, and increased the expression of Bcl-2 in H2O2 treated MLE-12 cells. Furthermore, Gpnmb knockdown could significantly reduce reactive oxygen species (ROS) generation and improve the mitochondrial membrane potential.Conclusion: The present study showed that knockdown of Gpnmb may protect against HLI by repressing mitochondrial-mediated apoptosis.
Collapse
Affiliation(s)
- Xiaoqin Wang
- Department of Pediatrics, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Song Qin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yingcong Ren
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Banghai Feng
- Department of Critical Care Medicine, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, China
| | - Junya Liu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Kun Yu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hong Yu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhenliang Liao
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hong Mei
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Mei Tan
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Pediatrics, Guizhou Children's Hospital, Zunyi, China
- Collaborative Innovation Center for Tissue Injury Repair and Regenerative Medicine of Zunyi Medical University, Zunyi, China
| |
Collapse
|
7
|
Qin S, Liu JY, Wang XQ, Feng BH, Ren YC, Zheng J, Yu K, Yu H, Li K, Zhu F, Chen M, Fu X, Chen T, Xing ZX, Mei H. ROS-mediated MAPK activation aggravates hyperoxia-induced acute lung injury by promoting apoptosis of type II alveolar epithelial cells via the STAT3/miR-21-5p axis. Mol Immunol 2023; 163:207-215. [PMID: 37839259 DOI: 10.1016/j.molimm.2023.09.016] [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: 03/30/2023] [Revised: 09/13/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023]
Abstract
Inhibition of type II alveolar epithelial (AE-II) cell apoptosis is a critical way to cure hyperoxia-induced acute lung injury (HALI). It has been reported that miR-21-5p could reduce H2O2-induced apoptosis in AE-II cells. However, the upstream molecular mechanism remains unclear. Herein, we established a cellular model of HALI by exposing AE-II cells to H2O2 treatment. It was shown that miR-21-5p alleviated H2O2-induced apoptosis in AE-II cells. ROS inhibition decreased apoptosis of H2O2-evoked AE-II cells via increasing miR-21-5p expression. In addition, ROS induced MAPK and STAT3 phosphorylation in H2O2-treated AE-II cells. MAPK inactivation reduces H2O2-triggered AE-II cell apoptosis. MAPK activation inhibits miR-21-5p expression by promoting STAT3 phosphorylation in H2O2-challenged AE-II cells. Furthermore, STAT3 activation eliminated MAPK deactivation-mediated inhibition on the apoptosis of AE-II cells under H2O2 condition. In conclusion, ROS-mediated MAPK activation promoted H2O2-triggered AE-II cell apoptosis by inhibiting miR-21-5p expression via STAT3 phosphorylation, providing novel targets for HALI treatment.
Collapse
Affiliation(s)
- Song Qin
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Jun-Ya Liu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Xiao-Qin Wang
- Department of Pediatric, The second affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Bang-Hai Feng
- Department of Critical Care Medicine, Zunyi Hospital of Traditional Chinese Medicine, Zunyi 563000, PR China
| | - Ying-Cong Ren
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Jie Zheng
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Kun Yu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Hong Yu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Kang Li
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Feng Zhu
- Department of Respiratory and Critical Care Medicine, The Fifth People's Hospital of Wuxi Affiliated to Jiangnan University, Wuxi 214016, PR China
| | - Miao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Xiaoyun Fu
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Tao Chen
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China
| | - Zhou-Xiong Xing
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China.
| | - Hong Mei
- Department of Critical Care Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, PR China.
| |
Collapse
|
8
|
Das S, Taylor K, Kozubek J, Sardell J, Gardner S. Genetic risk factors for ME/CFS identified using combinatorial analysis. J Transl Med 2022; 20:598. [PMID: 36517845 PMCID: PMC9749644 DOI: 10.1186/s12967-022-03815-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic disease that lacks known pathogenesis, distinctive diagnostic criteria, and effective treatment options. Understanding the genetic (and other) risk factors associated with the disease would begin to help to alleviate some of these issues for patients. METHODS We applied both GWAS and the PrecisionLife combinatorial analytics platform to analyze ME/CFS cohorts from UK Biobank, including the Pain Questionnaire cohort, in a case-control design with 1000 cycles of fully random permutation. Results from this study were supported by a series of replication and cohort comparison experiments, including use of disjoint Verbal Interview CFS, post-viral fatigue syndrome and fibromyalgia cohorts also derived from UK Biobank, and compared results for overlap and reproducibility. RESULTS Combinatorial analysis revealed 199 SNPs mapping to 14 genes that were significantly associated with 91% of the cases in the ME/CFS population. These SNPs were found to stratify by shared cases into 15 clusters (communities) made up of 84 high-order combinations of between 3 and 5 SNPs. p-values for these communities range from 2.3 × 10-10 to 1.6 × 10-72. Many of the genes identified are linked to the key cellular mechanisms hypothesized to underpin ME/CFS, including vulnerabilities to stress and/or infection, mitochondrial dysfunction, sleep disturbance and autoimmune development. We identified 3 of the critical SNPs replicated in the post-viral fatigue syndrome cohort and 2 SNPs replicated in the fibromyalgia cohort. We also noted similarities with genes associated with multiple sclerosis and long COVID, which share some symptoms and potentially a viral infection trigger with ME/CFS. CONCLUSIONS This study provides the first detailed genetic insights into the pathophysiological mechanisms underpinning ME/CFS and offers new approaches for better diagnosis and treatment of patients.
Collapse
Affiliation(s)
- Sayoni Das
- PrecisionLife Ltd, Long Hanborough, Oxford, UK
| | | | | | | | | |
Collapse
|
9
|
Soundararajan R, Hernández-Cuervo H, Stearns TM, Griswold AJ, Patil SS, Fukumoto J, Narala VR, Galam L, Lockey R, Kolliputi N. A-Kinase Anchor Protein 1 deficiency causes mitochondrial dysfunction in mouse model of hyperoxia induced acute lung injury. Front Pharmacol 2022; 13:980723. [PMID: 36263130 PMCID: PMC9574061 DOI: 10.3389/fphar.2022.980723] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Critically ill patients on supplemental oxygen therapy eventually develop acute lung injury (ALI). Reactive oxygen species (ROS) produced during ALI perturbs the mitochondrial dynamics resulting in cellular damage. Genetic deletion of the mitochondrial A-kinase anchoring protein 1 (Akap1) in mice resulted in mitochondrial damage, Endoplasmic reticulum (ER) stress, increased expression of mitophagy proteins and pro-inflammatory cytokines, exacerbating hyperoxia-induced Acute Lung Injury (HALI).Objective: Despite a strong causal link between mitochondrial dysfunction and HALI, the mechanisms governing the disease progression at the transcriptome level is unknown.Methods: In this study, RNA sequencing (RNA-seq) analysis was carried out using the lungs of Akap1 knockout (Akap1−/−) mice exposed to normoxia or 48 h of hyperoxia followed by quantitative real time PCR and Ingenuity pathway analysis (IPA). Western blot analysis assessed mitochondrial dysfunction, OXPHOS complex (I-V), apoptosis and antioxidant proteins. Mitochondrial enzymatic assays was used to measure the aconitase, fumarase, citrate synthase activities in isolated mitochondria from Akap1−/− vs. Wt mice exposed to hyperoxia.Results: Transcriptome analysis of Akap1−/− exposed to hyperoxia reveals increases in transcripts encoding electron transport chain (ETC) and tricarboxylic acid cycle (TCA) proteins. Ingenuity pathway analysis (IPA) shows enrichment of mitochondrial dysfunction and oxidative phosphorylation in Akap1−/− mice. Loss of AKAP1, coupled with oxidant injury, significantly decreases the activities of TCA enzymes. Mechanistically, a significant loss of dynamin-related protein 1 (Drp1) phosphorylation at the protein kinase A (PKA) site Serine 637 (Ser637), decreases in Akt phosphorylation at Serine 437 (Ser47) and increase in the expression of pro-apoptotic protein Bax indicate mitochondrial dysfunction. Heme oxygenase-1 (HO-1) levels significantly increased in CD68 positive alveolar macrophages in Akap1−/− lungs, suggesting a strong antioxidant response to hyperoxia.Conclusion: Overall these results suggest that AKAP1 overexpression and modulation of Drp1 phosphorylation at Ser637 is an important therapeutic strategy for acute lung injury.
Collapse
Affiliation(s)
- Ramani Soundararajan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Helena Hernández-Cuervo
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- University of South Florida, Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL, United States
| | | | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Sahebgowda Sidramagowda Patil
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jutaro Fukumoto
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | | | - Lakshmi Galam
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Richard Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
- University of South Florida, Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL, United States
- *Correspondence: Narasaiah Kolliputi,
| |
Collapse
|
10
|
Narala VR, Thimmana LV, Panati K, Kolliputi N. Nitrated fatty acid, 10-nitrooleate protects against hyperoxia-induced acute lung injury in mice. Int Immunopharmacol 2022; 109:108838. [PMID: 35561478 DOI: 10.1016/j.intimp.2022.108838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 11/19/2022]
Abstract
The antioxidant and anti-inflammatory effects of electrophilic nitrated fatty acid (NFA); 10-nitrooleate, have been reported. The present study investigated whether 10-nitrooleate has a protective role against hyperoxic-induced acute lung injury (HALI). Using a C57BL/6 mice model of HALI, we investigated the protective effect of 10-nitrooleate. C57BL/6 mice were administered with NFA intratracheally, exposed to hyperoxia for 48 h to induce HALI, and kept at room air for 24 h. Bronchoalveolar lavage (BAL) fluid and lung samples were collected after 24 h of post hyperoxia to analyze markers associated with HALI. Intratracheal (IT) and intraperitoneal (IP) administration of NFA notably attenuated hyperoxia-induced infiltration of inflammatory cells, alveolar-capillary leakage, upregulation of proinflammatory cytokine levels (IL-6 and TNFα) into the BAL fluid, and resolution of inflammation in the lung. Western blot analyses showed that 10-nitrooleate reduced the expression of the inflammatory transcription factor NFκB p65 subunit and increased antioxidant proteins HO-1 and NQO1 expression in the lung tissues compared to vehicle-treated animals. Moreover, 10-nitrooleate reversed the hyperoxia-induced expression of mitophagy-associated markers (PINK1 and p62/SQSTM1), thereby protecting the HALI/ acute respiratory distress syndrome (ARDS). IT and IP delivery of 10-nitrooleate reduces hyperoxia-induced ALI/ARDS by regulating the antioxidant pathways and restoring the mitochondrial homeostasis by regulating mitophagy. It is suggested that NFAs can be further evaluated as supplementary therapy for critically ill patients like COVID-19/ARDS.
Collapse
Affiliation(s)
| | - Lokesh V Thimmana
- Department of Zoology, Yogi Vemana University, Kadapa, 516 005, Andhra Pradesh, India
| | - Kalpana Panati
- Department of Biotechnology, Government College for Men, Kadapa, Andhra Pradesh, India
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
11
|
Sidramagowda Patil S, Soundararajan R, Fukumoto J, Breitzig M, Hernández-Cuervo H, Alleyn M, Lin M, Narala VR, Lockey R, Kolliputi N, Galam L. Mitochondrial Protein Akap1 Deletion Exacerbates Endoplasmic Reticulum Stress in Mice Exposed to Hyperoxia. Front Pharmacol 2022; 13:762840. [PMID: 35370705 PMCID: PMC8964370 DOI: 10.3389/fphar.2022.762840] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/27/2022] [Indexed: 01/02/2023] Open
Abstract
Acute lung injury (ALI) and its severe manifestation, acute respiratory distress syndrome (ARDS), are treated with high concentrations of supplementary oxygen. However, prolonged exposure to high oxygen concentrations stimulates the production of reactive oxygen species (ROS), which damages the mitochondria and accumulates misfolded proteins in the endoplasmic reticulum (ER). The mitochondrial protein A-kinase anchoring protein 1 (Akap1) is critical for mitochondrial homeostasis. It is known that Akap1 deficiency results in heart damage, neuronal development impairment, and mitochondrial malfunction in preclinical studies. Our laboratory recently revealed that deleting Akap1 increases the severity of hyperoxia-induced ALI in mice. To assess the role of Akap1 deletion in ER stress in lung injury, wild-type and Akap1−/− mice were exposed to hyperoxia for 48 h. This study indicates that Akap1−/− mice exposed to hyperoxia undergo ER stress, which is associated with an increased expression of BiP, JNK phosphorylation, eIF2α phosphorylation, ER stress-induced cell death, and autophagy. This work demonstrates that deleting Akap1 results in increased ER stress in the lungs of mice and that hyperoxia exacerbates ER stress-related consequences.
Collapse
Affiliation(s)
- Sahebgowda Sidramagowda Patil
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Ramani Soundararajan
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Jutaro Fukumoto
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Mason Breitzig
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States.,Washington University in St. Louis, Brown School, St. Louis, MO, United States
| | - Helena Hernández-Cuervo
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States.,University of South Florida, Department of Molecular Medicine, College of Medicine, Tampa, FL, United States
| | - Matthew Alleyn
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Muling Lin
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | | | - Richard Lockey
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| | - Narasaiah Kolliputi
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States.,University of South Florida, Department of Molecular Medicine, College of Medicine, Tampa, FL, United States
| | - Lakshmi Galam
- University of South Florida, Division of Allergy and Immunology, Department of Internal Medicine, College of Medicine, Tampa, FL, United States
| |
Collapse
|
12
|
Kong M, Wei D, Li X, Zhu X, Hong Z, Ni M, Wang Y, Dong A. The dynamic changes in autophagy activity and its role in lung injury after deep hypothermic circulatory arrest. J Cell Mol Med 2022; 26:1113-1127. [PMID: 35014165 PMCID: PMC8831962 DOI: 10.1111/jcmm.17165] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/22/2021] [Accepted: 12/09/2021] [Indexed: 12/27/2022] Open
Abstract
Deep hypothermic circulatory arrest (DHCA) can cause acute lung injury (ALI), and its pathogenesis mimics ischaemia/reperfusion (I/R) injury. Autophagy is also involved in lung I/R injury. The present study aimed to elucidate whether DHCA induces natural autophagy activation and its role in DHCA‐mediated lung injury. Here, rats were randomly assigned to the Sham or DHCA group. The sham group (n = 5) only received anaesthesia and air intubation. DHCA group rats underwent cardiopulmonary bypass (CPB) followed by the DHCA procedure. The rats were then sacrificed at 3, 6 and 24 h after the DHCA procedure (n = 5) to measure lung injury and autophagy activity. Chloroquine (CQ) was delivered to evaluate autophagic flux. DHCA caused lung injury, which was prominent 3–6 h after DHCA, as confirmed by histological examination and inflammatory cytokine quantification. Lung injury subsided at 24 h. Autophagy was suppressed 3 h but was exaggerated at 6 h. At both time points, autophagic flux appeared uninterrupted. To further assess the role of autophagy in DHCA‐mediated lung injury, the autophagy inducer rapamycin and its inhibitor 3‐methyladenine (3‐MA) were applied, and lung injury was reassessed. When rapamycin was administered at an early time point, lung injury worsened, whereas administration of 3‐MA at a late time point ameliorated lung injury, indicating that autophagy contributed to lung injury after DHCA. Our study presents a time course of lung injury following DHCA. Autophagy showed adaptive yet protective suppression 3 h after DHCA, as induction of autophagy caused worsening of lung tissue. In contrast, autophagy was exaggerated 6 h after DHCA, and autophagy inhibition attenuated DHCA‐mediated lung injury.
Collapse
Affiliation(s)
- Minjian Kong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Dongdong Wei
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xuebiao Li
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xian Zhu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ze Hong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Ni
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Wang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Aiqiang Dong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
13
|
Soundararajan R, Varanasi SM, Patil SS, Srinivas S, Hernández-Cuervo H, Czachor A, Bulkhi A, Fukumoto J, Galam L, Lockey RF, Kolliputi N. Lung fibrosis is induced in ADAR2 overexpressing mice via HuR-induced CTGF signaling. FASEB J 2022; 36:e22143. [PMID: 34985777 PMCID: PMC10395739 DOI: 10.1096/fj.202101511r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 11/11/2022]
Abstract
Adenosine deaminase acting on RNA 2 (ADAR2), an RNA editing enzyme is involved in a site-selective modification of adenosine (A) to inosine (I) in double-stranded RNA (dsRNA). Its role in the lungs is unknown. The phenotypic characterization of Adarb1 mice that lacked ADAR2 auto-regulation due to the deletion of editing complementary sequence (ΔECS mice) determined the functional role of ADAR2 in the lungs. ADAR2 protein expression increased in the ΔECS mice. These mice display immune cell infiltration and alveolar disorganization. The lung wet by dry ratio indicates there is no lung edema in ΔECS mice. Bronchoalveolar lavage (BAL) analysis of ΔECS mice reveals a significant increase in neutrophils. Interestingly, ΔECS mice spontaneously develop lung fibrosis as indicated by Sirius red staining of collagen fibers in the lung sections and a significant increase in hydroxyproline level in their lungs. ADAR2 expression increased significantly in a bleomycin mouse model, implicating a role of ADAR2 in lung fibrosis. Furthermore, there is a likely possibility that the genetically modified ΔECS mice does not model the physiological or pathophysiological process of lung fibrosis. Nevertheless, this model is useful in interrogating the role of ADAR2 in the lungs. The Ctgf mRNA and connective tissue growth factor (CTGF) protein significantly increased in ΔECS lungs and occurs in bronchial epithelial cells. There is a significant increase in Human antigen R (ELAVL1; HuR) protein levels in ΔECS lungs and suggests a role in stabilizing Ctgf mRNA. Lung mechanics such as total respiratory resistance, Newtonian resistance and tissue damping were increased, whereas inspiratory capacity was decreased in the ΔECS mice. Taken together, these data indicate that overexpression of ADAR2 causes spontaneous lung fibrosis via HuR-mediated CTGF signaling and implicate a role for ADAR2 auto-regulation in lung homeostasis. The identification of ADAR2 target genes in ΔECS mice would facilitate a mechanistic understanding of the role of ADAR2 in the lungs and provide a therapeutic strategy for lung fibrosis.
Collapse
Affiliation(s)
- Ramani Soundararajan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Sai Manasa Varanasi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Sahebgowda Sidramagowda Patil
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Sriraja Srinivas
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA.,Department of Drug Discovery and Development, Auburn University, Auburn, Alabama, USA
| | - Helena Hernández-Cuervo
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Alexander Czachor
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA.,Department of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Adeeb Bulkhi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA.,Department of Internal Medicine, College of Medicine, Umm Al Qura University, Makkah, Saudi Arabia
| | - Jutaro Fukumoto
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Lakshmi Galam
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Richard F Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
14
|
Jia Q, Yang Y, Chen X, Yao S, Hu Z. Emerging roles of mechanosensitive ion channels in acute lung injury/acute respiratory distress syndrome. Respir Res 2022; 23:366. [PMID: 36539808 PMCID: PMC9764320 DOI: 10.1186/s12931-022-02303-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a devastating respiratory disorder with high rates of mortality and morbidity, but the detailed underlying mechanisms of ALI/ARDS remain largely unknown. Mechanosensitive ion channels (MSCs), including epithelial sodium channel (ENaC), Piezo channels, transient receptor potential channels (TRPs), and two-pore domain potassium ion (K2P) channels, are highly expressed in lung tissues, and the activity of these MSCs can be modulated by mechanical forces (e.g., mechanical ventilation) and other stimuli (e.g., LPS, hyperoxia). Dysfunction of MSCs has been found in various types of ALI/ARDS, and MSCs play a key role in regulating alveolar fluid clearance, alveolar epithelial/endothelial barrier function, the inflammatory response and surfactant secretion in ALI/ARDS lungs. Targeting MSCs exerts therapeutic effects in the treatment of ALI/ARDS. In this review, we summarize the structure and functions of several well-recognized MSCs, the role of MSCs in the pathogenesis of ALI/ARDS and recent advances in the pharmacological and molecular modulation of MSCs in the treatment of ALI/ARDS. According to the current literature, targeting MSCs might be a very promising therapeutic approach against ALI/ARDS.
Collapse
Affiliation(s)
- Qi Jia
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyi Yang
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangdong Chen
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanglong Yao
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Hu
- grid.33199.310000 0004 0368 7223Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Wang C, Li YH, Yang ZT, Cheng NT, Tang HX, Xu M. The function and mechanism of microRNA-92a-3p in lipopolysaccharide-induced acute lung injury. Immunopharmacol Immunotoxicol 2021; 44:47-57. [PMID: 34783628 DOI: 10.1080/08923973.2021.2001497] [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] [Indexed: 01/07/2023]
Abstract
OBJECTIVES Sepsis-associated acute lung injury (ALI) is a clinically severe respiratory disorder and remains the leading cause of multiple organ failure and mortality. Herein, we used lipopolysaccharide (LPS) to generate sepsis-induced ALI and try to explore the role and mechanism of microRNA-92a-3p (miR-92a-3p) in this process. METHODS Mice were intravenously injected with miR-92a-3p agomir, antagomir and negative controls for 3 consecutive days and then were intratracheally instillated by LPS (5 mg/kg) for 12 h. To knock down the endogenous A-kinase anchoring protein 1 (AKAP1), mice were intratracheally injected with recombinant adenovirus carrying the short hairpin RNA targeting AKAP1 (shAkap1) at 1 week before LPS administration. RESULTS miR-92a-3p level was significantly upregulated in the lungs by LPS injection. miR-92a-3p antagomir reduced LPS-induced intrapulmonary inflammation and oxidative stress, thereby preventing pulmonary injury and dysfunction. In contrast, miR-92a-3p agomir aggravated LPS-induced intrapulmonary inflammation, oxidative stress, pulmonary injury and dysfunction. Moreover, we reported that AKAP1 upregulation was required for the beneficial effects of miR-92a-3p antagomir, and that AKAP1 knockdown completely abolished the anti-inflammatory and antioxidant capacities of miR-92a-3p antagomir. CONCLUSION Our data identify that miR-92a-3p modulates LPS-induced intrapulmonary inflammation, oxidative stress and ALI via AKAP1 in mice.
Collapse
Affiliation(s)
- Cong Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yang-Hao Li
- Department of Thoracic Surgery, Huangmei People's Hospital, Huanggang, China
| | - Ze-Tian Yang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ni-Tao Cheng
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - He-Xiao Tang
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ming Xu
- Department of Thoracic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
16
|
Aboushanab SA, El-Far AH, Narala VR, Ragab RF, Kovaleva EG. Potential therapeutic interventions of plant-derived isoflavones against acute lung injury. Int Immunopharmacol 2021; 101:108204. [PMID: 34619497 DOI: 10.1016/j.intimp.2021.108204] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/19/2021] [Accepted: 09/25/2021] [Indexed: 12/24/2022]
Abstract
Acute lung injury (ALI) is a life-threatening syndrome that possibly leads to high morbidity and mortality as no therapy exists. Several natural ingredients with negligible adverse effects have recently been investigated to possibly inhibit the inflammatory pathways associated with ALI at the molecular level. Isoflavones, as phytoestrogenic compounds, are naturally occurring bioactive compounds that represent the most abundant category of plant polyphenols (Leguminosae family). A broad range of therapeutic activities of isoflavones, including antioxidants, chemopreventive, anti-inflammatory, antiallergic and antibacterial potentials, have been extensively documented in the literature. Our review exclusively focuses on the possible anti-inflammatory, antioxidant role of botanicals'-derived isoflavones against ALI and their immunomodulatory effect in experimentally induced ALI. Despite the limited scope covering their molecular mechanisms, isoflavones substantially contributed to protecting from ALI via inhibiting toll-like receptor 4 (TLR4)/Myd88/NF-κB pathway and subsequent cytokines, chemokines, and adherent proteins. Nonetheless, future research is suggested to fill the gap in elucidating the protective roles of isoflavones to alleviate ALI concerning antioxidant potentials, inhibition of the inflammatory pathways, and associated molecular mechanisms.
Collapse
Affiliation(s)
- Saied A Aboushanab
- Institute of Chemical Engineering, Ural Federal University named after the First President of Russia B. N. Yeltsin, 620002, 19 Mira Yekaterinburg, Russia.
| | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt; Scientific Chair of Yousef Abdullatif Jameel of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
| | | | - Rokia F Ragab
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt.
| | - Elena G Kovaleva
- Institute of Chemical Engineering, Ural Federal University named after the First President of Russia B. N. Yeltsin, 620002, 19 Mira Yekaterinburg, Russia.
| |
Collapse
|
17
|
Sang Y, Liu J, Li X, Zhou G, Zhang Y, Gao L, Zhao Y, Zhou X. The effect of SiNPs on DNA methylation of genome in mouse spermatocytes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:43684-43697. [PMID: 33840017 DOI: 10.1007/s11356-021-13459-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 03/11/2021] [Indexed: 06/12/2023]
Abstract
Silica nanoparticles (SiNPs), which are the main inorganic components of atmospheric particulate matter, have been proved to have certain male reproductive toxicity in previous studies. Spermatogenesis involves complex epigenetic regulation, but it is still unclear if SiNPs exposure will interfere with the DNA methylation patterns in mouse spermatocytes. The present study was designed to investigate the effects of SiNPs on DNA methylation in the mouse spermatocyte GC-2spd(ts). GC-2 cells were treated with 0 and 20 μg/mL SiNPs for 24 h. MeDIP-seq assay was then performed to analyze the differentially methylated genes related to spermatogenesis. The results showed that SiNPs induced extensive methylation changes in the genome of GC-2 cells, and 24a total of 428 hyper-methylated genes and 398 hypo-methylated genes were identified. Gene Ontology and pathway analysis showed that differential DNA methylation induced by SiNPs was probably involved with abnormal transcription and translation, mitochondrial damage, and cell apoptosis. Results from qRT-PCR verification showed that the expression of spermatogenesis-related genes Akap1, Crem, Spz1, and Tex11 were dysregulated by SiNPs exposure, which was consistent with the MeDIP-seq assay. In general, this study suggested that SiNPs caused genome-wide DNA methylation changes in GC-2 cells, providing valuable reference for the future epigenetic studies in SiNPs-induced male reproductive toxicity.
Collapse
Affiliation(s)
- Yujian Sang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Jianhui Liu
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Xiangyang Li
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Guiqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yue Zhang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Leqiang Gao
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yanzhi Zhao
- Yanjing Medical College, Capital Medical University, Beijing, 100069, China.
| | - Xianqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
18
|
Pao HP, Liao WI, Tang SE, Wu SY, Huang KL, Chu SJ. Suppression of Endoplasmic Reticulum Stress by 4-PBA Protects Against Hyperoxia-Induced Acute Lung Injury via Up-Regulating Claudin-4 Expression. Front Immunol 2021; 12:674316. [PMID: 34122432 PMCID: PMC8194262 DOI: 10.3389/fimmu.2021.674316] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
Endoplasmic reticulum (ER) stress that disrupts ER function can occur in response to a wide variety of cellular stress factors leads to the accumulation of unfolded and misfolded proteins in the ER. Many studies have shown that ER stress amplified inflammatory reactions and was involved in various inflammatory diseases. However, little is known regarding the role of ER stress in hyperoxia-induced acute lung injury (HALI). This study investigated the influence of ER stress inhibitor, 4-phenyl butyric acid (4-PBA), in mice with HALI. Treatment with 4-PBA in the hyperoxia groups significantly prolonged the survival, decreased lung edema, and reduced the levels of inflammatory mediators, lactate dehydrogenase, and protein in bronchoalveolar lavage fluid, and increased claudin-4 protein expression in lung tissue. Moreover, 4-PBA reduced the ER stress-related protein expression, NF-κB activation, and apoptosis in the lung tissue. In in vitro study, 4-PBA also exerted a similar effect in hyperoxia-exposed mouse lung epithelial cells (MLE-12). However, when claudin-4 siRNA was administrated in mice and MLE-12 cells, the protective effect of 4-PBA was abrogated. These results suggested that 4-PBA protected against hyperoxia-induced ALI via enhancing claudin-4 expression.
Collapse
Affiliation(s)
- Hsin-Ping Pao
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wen-I Liao
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-En Tang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.,Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Yu Wu
- Institute of Aerospace and Undersea Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Kun-Lun Huang
- The Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shi-Jye Chu
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
19
|
Xuefei Y, Xinyi Z, Qing C, Dan Z, Ziyun L, Hejuan Z, Xindong X, Jianhua F. Effects of Hyperoxia on Mitochondrial Homeostasis: Are Mitochondria the Hub for Bronchopulmonary Dysplasia? Front Cell Dev Biol 2021; 9:642717. [PMID: 33996802 PMCID: PMC8120003 DOI: 10.3389/fcell.2021.642717] [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: 12/16/2020] [Accepted: 04/12/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are involved in energy metabolism and redox reactions in the cell. Emerging data indicate that mitochondria play an essential role in physiological and pathological processes of neonatal lung development. Mitochondrial damage due to exposure to high concentrations of oxygen is an indeed important factor for simplification of lung structure and development of bronchopulmonary dysplasia (BPD), as reported in humans and rodent models. Here, we comprehensively review research that have determined the effects of oxygen environment on alveolar development and morphology, summarize changes in mitochondria under high oxygen concentrations, and discuss several mitochondrial mechanisms that may affect cell plasticity and their effects on BPD. Thus, the pathophysiological effects of mitochondria may provide insights into targeted mitochondrial and BPD therapy.
Collapse
Affiliation(s)
- Yu Xuefei
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Zhao Xinyi
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Cai Qing
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Zhang Dan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Liu Ziyun
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Zheng Hejuan
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Xue Xindong
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| | - Fu Jianhua
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang City, China
| |
Collapse
|
20
|
Amarelle L, Quintela L, Hurtado J, Malacrida L. Hyperoxia and Lungs: What We Have Learned From Animal Models. Front Med (Lausanne) 2021; 8:606678. [PMID: 33768102 PMCID: PMC7985075 DOI: 10.3389/fmed.2021.606678] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Although oxygen (O2) is essential for aerobic life, it can also be an important source of cellular damage. Supra-physiological levels of O2 determine toxicity due to exacerbated reactive oxygen species (ROS) production, impairing the homeostatic balance of several cellular processes. Furthermore, injured cells activate inflammation cascades, amplifying the tissue damage. The lung is the first (but not the only) organ affected by this condition. Critically ill patients are often exposed to several insults, such as mechanical ventilation, infections, hypo-perfusion, systemic inflammation, and drug toxicity. In this scenario, it is not easy to dissect the effect of oxygen toxicity. Translational investigations with animal models are essential to explore injuring stimuli in controlled experimental conditions, and are milestones in understanding pathological mechanisms and developing therapeutic strategies. Animal models can resemble what happens in critical care or anesthesia patients under mechanical ventilation and hyperoxia, but are also critical to explore the effect of O2 on lung development and the role of hyperoxic damage on bronchopulmonary dysplasia. Here, we set out to review the hyperoxia effects on lung pathology, contributing to the field by describing and analyzing animal experimentation's main aspects and its implications on human lung diseases.
Collapse
Affiliation(s)
- Luciano Amarelle
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Lucía Quintela
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Javier Hurtado
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay
| | - Leonel Malacrida
- Department of Pathophysiology, Hospital de Clínicas, School of Medicine, Universidad de la República, Montevideo, Uruguay.,Advanced Bioimaging Unit, Institut Pasteur Montevideo and Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
21
|
Ji L, Zhao Y, He L, Zhao J, Gao T, Liu F, Qi B, Kang F, Wang G, Zhao Y, Guo H, He Y, Li F, Huang Q, Xing J. AKAP1 Deficiency Attenuates Diet-Induced Obesity and Insulin Resistance by Promoting Fatty Acid Oxidation and Thermogenesis in Brown Adipocytes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002794. [PMID: 33747723 PMCID: PMC7967052 DOI: 10.1002/advs.202002794] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/20/2020] [Indexed: 05/06/2023]
Abstract
Altering the balance between energy intake and expenditure is a major strategy for treating obesity. Nonetheless, despite the progression in antiobesity drugs on appetite suppression, therapies aimed at increasing energy expenditure are limited. Here, knockout ofAKAP1, a signaling hub on outer mitochondrial membrane, renders mice resistant to diet-induced obesity.AKAP1 knockout significantly enhances energy expenditure and thermogenesis in brown adipose tissues (BATs) of obese mice. Restoring AKAP1 expression in BAT clearly reverses the beneficial antiobesity effect in AKAP1-/- mice. Mechanistically, AKAP1 remarkably decreases fatty acid β-oxidation (FAO) by phosphorylating ACSL1 to inhibit its activity in a protein-kinase-A-dependent manner and thus inhibits thermogenesis in brown adipocytes. Importantly, AKAP1 peptide inhibitor effectively alleviates diet-induced obesity and insulin resistance. Altogether, the findings demonstrate that AKAP1 functions as a brake of FAO to promote diet-induced obesity, which may be used as a potential therapeutic target for obesity.
Collapse
Affiliation(s)
- Lele Ji
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
- National Demonstration Center for Experimental Preclinical Medicine EducationFourth Military Medical UniversityXi'anShaanxi710032China
| | - Ya Zhao
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
- Laboratory Animal CenterFourth Military Medical UniversityXi'anShaanxi710032China
| | - Linjie He
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
| | - Jing Zhao
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
| | - Tian Gao
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
| | - Fengzhou Liu
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Bingchao Qi
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Fei Kang
- Department of Nuclear MedicineXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Gang Wang
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
| | - Yilin Zhao
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
| | - Haitao Guo
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
| | - Yuanfang He
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
| | - Fei Li
- Department of CardiologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Qichao Huang
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology and Department of Physiology and PathophysiologyFourth Military Medical UniversityXi'anShaanxi710032China
| |
Collapse
|
22
|
Snyder RJ, Kleeberger SR. Role of Mitochondrial DNA in Inflammatory Airway Diseases. Compr Physiol 2021; 11:1485-1499. [PMID: 33577124 DOI: 10.1002/cphy.c200010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The mitochondrial genome is a small, circular, and highly conserved piece of DNA which encodes only 13 protein subunits yet is vital for electron transport in the mitochondrion and, therefore, vital for the existence of multicellular life on Earth. Despite this importance, mitochondrial DNA (mtDNA) is located in one of the least-protected areas of the cell, exposing it to high concentrations of intracellular reactive oxygen species (ROS) and threat from exogenous substances and pathogens. Until recently, the quality control mechanisms which ensured the stability of the nuclear genome were thought to be minimal or nonexistent in the mitochondria, and the thousands of redundant copies of mtDNA in each cell were believed to be the primary mechanism of protecting these genes. However, a vast network of mechanisms has been discovered that repair mtDNA lesions, replace and recycle mitochondrial chromosomes, and conduct alternate RNA processing for previously undescribed mitochondrial proteins. New mtDNA/RNA-dependent signaling pathways reveal a mostly undiscovered biochemical landscape in which the mitochondria interface with their host cells/organisms. As the myriad ways in which the function of the mitochondrial genome can affect human health have become increasingly apparent, the use of mitogenomic biomarkers (such as copy number and heteroplasmy) as toxicological endpoints has become more widely accepted. In this article, we examine several pathologies of human airway epithelium, including particle exposures, inflammatory diseases, and hyperoxia, and discuss the role of mitochondrial genotoxicity in the pathogenesis and/or exacerbation of these conditions. © 2021 American Physiological Society. Compr Physiol 11:1485-1499, 2021.
Collapse
Affiliation(s)
- Ryan J Snyder
- National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| | - Steven R Kleeberger
- National Institute of Environmental Health Sciences, NIH, Durham, North Carolina, USA
| |
Collapse
|
23
|
Derangula M, Panati K, Narala VR. Biochanin A Ameliorates Ovalbumin-induced Airway Inflammation through Peroxisome Proliferator-Activated Receptor-Gamma in a Mouse Model. Endocr Metab Immune Disord Drug Targets 2021; 21:145-155. [PMID: 32359341 DOI: 10.2174/1871530320666200503051609] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/09/2020] [Accepted: 04/01/2020] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Asthma is an inflammatory airway disease affecting most of the population in the world. The current medication for asthma relieves airway inflammation but it has serious adverse effects. Biochanin A (BCA), a phytoestrogen, is an active component present in red clover, alfalfa, soy having anti-oxidant and anti-inflammatory properties. BCA was identified as a natural activator of peroxisome proliferator-activated receptor-gamma (PPARγ). METHODS The study aims to evaluate the effects of BCA in ovalbumin (OVA)-induced murine model of asthma and to study the role of PPARγ. RESULTS We found that BCA administration reduced the severity of murine allergic asthma as evidenced histologically, and measurement of allergen-specific IgE levels in serum as well as in BAL fluid. BCA also reversed the elevated levels of inflammatory cytokines, cell infiltration, protein leakage into the airways and expression of hemoxygenase-1 in OVA-induced lungs. Further, we confirmed that BCA mediated inhibitory effects are mediated through PPARγ as assessed by treatment with PPARγ antagonist GW9662. CONCLUSION Our results suggest that BCA is efficacious in a preclinical model of asthma and may have the potential for the treatment of asthma in humans.
Collapse
Affiliation(s)
- Madhavi Derangula
- Department of Zoology, Yogi Vemana University, Kadapa, 516 005, Andhra Pradesh, India
| | - Kalpana Panati
- Department of Biotechnology, Government College for Men, Kadapa, Andhra Pradesh, India
| | - Venkata R Narala
- Department of Zoology, Yogi Vemana University, Kadapa, 516 005, Andhra Pradesh, India
| |
Collapse
|
24
|
Sidramagowda Patil S, Hernández-Cuervo H, Fukumoto J, Krishnamurthy S, Lin M, Alleyn M, Breitzig M, Narala VR, Soundararajan R, Lockey RF, Kolliputi N, Galam L. Alda-1 Attenuates Hyperoxia-Induced Acute Lung Injury in Mice. Front Pharmacol 2021; 11:597942. [PMID: 33597876 PMCID: PMC7883597 DOI: 10.3389/fphar.2020.597942] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/19/2020] [Indexed: 12/31/2022] Open
Abstract
Acute lung injury (ALI), a milder form of acute respiratory distress syndrome (ARDS), is a leading cause of mortality in older adults with an increasing prevalence. Oxygen therapy, is a common treatment for ALI, involving exposure to a high concentration of oxygen. Unfortunately, hyperoxia induces the formation of reactive oxygen species which can cause an increase in 4-HNE (4-hydroxy 2 nonenal), a toxic byproduct of lipid peroxidation. Mitochondrial aldehyde dehydrogenase 2 (ALDH2) serves as an endogenous shield against oxidative stress-mediated damage by clearing 4-HNE. Alda-1 [(N-(1, 3 benzodioxol-5-ylmethyl)-2, 6- dichloro-benzamide)], a small molecular activator of ALDH2, protects against reactive oxygen species-mediated oxidative stress by promoting ALDH2 activity. As a result, Alda-1 shields against ischemic reperfusion injury, heart failure, stroke, and myocardial infarction. However, the mechanisms of Alda-1 in hyperoxia-induced ALI remains unclear. C57BL/6 mice implanted with Alzet pumps received Alda-1 in a sustained fashion while being exposed to hyperoxia for 48 h. The mice displayed suppressed immune cell infiltration, decreased protein leakage and alveolar permeability compared to controls. Mechanistic analysis shows that mice pretreated with Alda-1 also experience decreased oxidative stress and enhanced levels of p-Akt and mTOR pathway associated proteins. These results show that continuous delivery of Alda-1 protects against hyperoxia-induced lung injury in mice.
Collapse
Affiliation(s)
- Sahebgowda Sidramagowda Patil
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Helena Hernández-Cuervo
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jutaro Fukumoto
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Sudarshan Krishnamurthy
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Muling Lin
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Matthew Alleyn
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Mason Breitzig
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Brown School, Washington University, St. Louis, MO, United States
| | | | - Ramani Soundararajan
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Richard F Lockey
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Narasaiah Kolliputi
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.,Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Lakshmi Galam
- Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| |
Collapse
|
25
|
miR-21-5p Suppresses Mitophagy to Alleviate Hyperoxia-Induced Acute Lung Injury by Directly Targeting PGAM5. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4807254. [PMID: 33681349 PMCID: PMC7907750 DOI: 10.1155/2020/4807254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/05/2020] [Indexed: 11/17/2022]
Abstract
Hyperoxia-induced acute lung injury (HALI) is a severe side effect of refractory hypoxemia treatment, for which no effective therapeutic strategy is available. Here, we found that the lung miR-21-5p level was significantly decreased in the rats subjected to hyperoxia. Further, we presented evidence that miR-21-5p was a crucial regulator of mitophagy and mitochondrial dysfunction. Moreover, it proved that miR-21-5p regulated hyperoxia-induced mitophagy and mitochondrial dysfunction by directly binding to the target gene PGAM5. In conclusion, for the first time, we found that miR-21-5p could directly suppress mitophagy and mitochondrial damage during HALI formation.
Collapse
|
26
|
Alda-1 attenuates hyperoxia-induced mitochondrial dysfunction in lung vascular endothelial cells. Aging (Albany NY) 2020; 11:3909-3918. [PMID: 31209184 PMCID: PMC6628993 DOI: 10.18632/aging.102012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/31/2019] [Indexed: 12/19/2022]
Abstract
Acute lung injury (ALI) is a major cause of morbidity and mortality worldwide, especially in aged populations. Mitochondrial damage is one of the key features of ALI. Hyperoxia-induced lung injury model in mice has been widely used for ALI study because it features many ALI phenotypes including, but not limited to, mitochondrial and vascular endothelial cell damage. Recently, accumulating evidence has shown that mitochondrial aldehyde dehydrogenase 2 (ALDH2) has a protective effect against oxidative stress mediated cell damage in epithelial cells. However, it is not known whether ALDH2 protects against oxidative stress in vascular endothelial cells. In this current study, we attempted to find the capacity of Alda-1 [(N-(1,3benzodioxol-5-ylmethyl)-2,6- dichloro-benzamide), an ALDH2 activator] to protect against oxidative stress in human microvascular endothelial cells (HMVEC). HMVEC pretreated with Alda-1 prior to hyperoxic exposure vs non-treated controls showed i) lower 4-hydroxynonenal (4-HNE) levels, ii) significantly decreased expressions of Bax and Cytochrome C, iii) partially restored activity and expression of ALDH2 and iv) significantly improved mitochondrial membrane potential. These results suggest that ALDH2 protein in lung vascular endothelial cells is a promising therapeutic target for the treatment of ALI and that Alda-1 is a potential treatment option.
Collapse
|
27
|
Qasim H, McConnell BK. AKAP12 Signaling Complex: Impacts of Compartmentalizing cAMP-Dependent Signaling Pathways in the Heart and Various Signaling Systems. J Am Heart Assoc 2020; 9:e016615. [PMID: 32573313 PMCID: PMC7670535 DOI: 10.1161/jaha.120.016615] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heart failure is a complex clinical syndrome, represented as an impairment in ventricular filling and myocardial blood ejection. As such, heart failure is one of the leading causes of death in the United States. With a mortality rate of 1 per 8 individuals and a prevalence of 6.2 million Americans, it has been projected that heart failure prevalence will increase by 46% by 2030. Cardiac remodeling (a general determinant of heart failure) is regulated by an extensive network of intertwined intracellular signaling pathways. The ability of signalosomes (molecular signaling complexes) to compartmentalize several cellular pathways has been recently established. These signalosome signaling complexes provide an additional level of specificity to general signaling pathways by regulating the association of upstream signals with downstream effector molecules. In cardiac myocytes, the AKAP12 (A-kinase anchoring protein 12) scaffolds a large signalosome that orchestrates spatiotemporal signaling through stabilizing pools of phosphatases and kinases. Predominantly upon β-AR (β2-adrenergic-receptor) stimulation, the AKAP12 signalosome is recruited near the plasma membrane and binds tightly to β-AR. Thus, one major function of AKAP12 is compartmentalizing PKA (protein kinase A) signaling near the plasma membrane. In addition, it is involved in regulating desensitization, downregulation, and recycling of β-AR. In this review, the critical roles of AKAP12 as a scaffold protein in mediating signaling downstream GPCRs (G protein-coupled receptor) are discussed with an emphasis on its reported and potential roles in cardiovascular disease initiation and progression.
Collapse
Affiliation(s)
- Hanan Qasim
- Department of Pharmacological and Pharmaceutical SciencesCollege of PharmacyUniversity of HoustonTX
| | - Bradley K. McConnell
- Department of Pharmacological and Pharmaceutical SciencesCollege of PharmacyUniversity of HoustonTX
| |
Collapse
|
28
|
Panati K, Thimmana LV, Narala VR. Electrophilic nitrated fatty acids are potential therapeutic candidates for inflammatory and fibrotic lung diseases. Nitric Oxide 2020; 102:28-38. [PMID: 32574817 DOI: 10.1016/j.niox.2020.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022]
Abstract
Several types of exposures can cause acute or chronic inflammatory reactions in the lungs often leading to asthma, pulmonary fibrosis, chronic obstructive pulmonary disease (COPD), acute lung injury, lung cancer, and other deleterious health outcomes. Current therapy, with inhaled or oral glucocorticoids, successfully targets inflammation but also produces adverse effects that limit their enthusiastic use. Accordingly, the need remains for interventions that are safer and more effective. Nitrated fatty acids (NFAs) are highly electrophilic and are produced endogenously by non-enzymatic reactions of nitric oxide with conjugated unsaturated fatty acids. The literature indicates that NFAs are detected in humans at the nanomolar range and are produced more robustly under inflammatory conditions. Recent studies on novel NFAs report antiinflammatory, antioxidant, and antifibrotic effects, while also acting as partial agonists of peroxisome proliferator-activated receptor-gamma (PPAR-γ). Furthermore, these functions of NFAs occur via reversible electrophilic alkylation of cysteine residues and regulation of antiinflammatory, antioxidant signaling through modulation of transcription factors, including nuclear factor E2-related factor 2 (Nrf2), PPAR-γ, and NF-κB. Here, we review and update the role of NFA signaling mechanisms and their therapeutic potential in various lung diseases. As NFAs display strong electrophilic interaction with multimechanistic pathways, they can be considered promising drug candidates for challenging lung diseases.
Collapse
Affiliation(s)
- Kalpana Panati
- Department of Biotechnology, Government College for Men, Kadapa, A.P, India
| | - Lokesh V Thimmana
- Department of Zoology, Yogi Vemana University, Kadapa, 516 005, A.P, India
| | | |
Collapse
|
29
|
Zheng D, Liu D, Kuang Y, Xu J, Xu G, Tai Q. Toll-like receptor 7 deficiency mitigates hyperoxia-induced acute lung injury in mice. Biomed Pharmacother 2020; 129:110345. [PMID: 32535385 DOI: 10.1016/j.biopha.2020.110345] [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] [Received: 03/18/2020] [Revised: 05/10/2020] [Accepted: 05/30/2020] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION Toll-like receptor (TLR) 7 is an important mediator in inflammation. However, its role in hyperoxia-induced acute lung injury (HALI) remains to be elucidated. METHODS C57BL/6 wild-type and C57BL/6 background TLR 7 deficiency mice were exposed to hyperoxia to stimulate HALI in airtight cages. Animals were sacrificed at 72 h post hyperoxia or room air exposure. Lung injury indicators were measured. Moreover, soluble epoxide hydrolase (sEH) activity was detected by a 14, 15-EET/DHET ELISA kit. Activation of activator protein (AP)-1 and nuclear factor kappa-B (NF-κB) was detected with enzyme linked immunosorbent assay kits. RESULTS Our data revealed that pulmonary histological assay and wet to dry weight ratio, myeloperoxidase and malondialdehyde activity were reduced in TLR 7 deficiency mice compared with wild-type mice. Moreover, hyperoxia-caused elevation of sEH activity was reduced in TLR 7 deficiency mice. Transcription factors AP-1 activation was significantly inhibited in TLR 7 deficiency mice compared with wild-type mice. Similarly, the activation of NF-κB was reduced in TLR 7 deficiency mice. Tumor necrosis factor-α and interleukin-1β, potent proinflammatory cytokines, were reduced in TLR 7 deficiency mice. CONCLUSION TLR 7 deficiency is associated with inhibition of inflammation in HALI in mice.
Collapse
Affiliation(s)
- Donghua Zheng
- Department Of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China
| | - Dawei Liu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China
| | - Yukun Kuang
- The Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China
| | - Jinghong Xu
- Department Of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China
| | - Guixing Xu
- Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, No. 58 Zhongshan Er Road, Guangzhou, GuangDong, 510000, China.
| | - Qiang Tai
- Department Of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, GuangDong, 510000, China.
| |
Collapse
|
30
|
Qi B, He L, Zhao Y, Zhang L, He Y, Li J, Li C, Zhang B, Huang Q, Xing J, Li F, Li Y, Ji L. Akap1 deficiency exacerbates diabetic cardiomyopathy in mice by NDUFS1-mediated mitochondrial dysfunction and apoptosis. Diabetologia 2020; 63:1072-1087. [PMID: 32072193 DOI: 10.1007/s00125-020-05103-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/06/2020] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS Diabetic cardiomyopathy, characterised by increased oxidative damage and mitochondrial dysfunction, contributes to the increased risk of heart failure in individuals with diabetes. Considering that A-kinase anchoring protein 121 (AKAP1) is localised in the mitochondrial outer membrane and plays key roles in the regulation of mitochondrial function, this study aimed to investigate the role of AKAP1 in diabetic cardiomyopathy and explore its underlying mechanisms. METHODS Loss- and gain-of-function approaches were used to investigate the role of AKAP1 in diabetic cardiomyopathy. Streptozotocin (STZ) was injected into Akap1-knockout (Akap1-KO) mice and their wild-type (WT) littermates to induce diabetes. In addition, primary neonatal cardiomyocytes treated with high glucose were used as a cell model of diabetes. Cardiac function was assessed with echocardiography. Akap1 overexpression was conducted by injecting adeno-associated virus 9 carrying Akap1 (AAV9-Akap1). LC-MS/MS analysis and functional experiments were used to explore underlying molecular mechanisms. RESULTS AKAP1 was downregulated in the hearts of STZ-induced diabetic mouse models. Akap1-KO significantly aggravated cardiac dysfunction in the STZ-treated diabetic mice when compared with WT diabetic littermates, as evidenced by the left ventricular ejection fraction (LVEF; STZ-treated WT mice [WT/STZ] vs STZ-treated Akap1-KO mice [KO/STZ], 51.6% vs 41.6%). Mechanistically, Akap1 deficiency impaired mitochondrial respiratory function characterised by reduced ATP production. Additionally, Akap1 deficiency increased cardiomyocyte apoptosis via enhanced mitochondrial reactive oxygen species (ROS) production. Furthermore, immunoprecipitation and mass spectrometry analysis indicated that AKAP1 interacted with the NADH-ubiquinone oxidoreductase 75 kDa subunit (NDUFS1). Specifically, Akap1 deficiency inhibited complex I activity by preventing translocation of NDUFS1 from the cytosol to mitochondria. Akap1 deficiency was also related to decreased ATP production and enhanced mitochondrial ROS-related apoptosis. In contrast, restoration of AKAP1 expression in the hearts of STZ-treated diabetic mice promoted translocation of NDUFS1 to mitochondria and alleviated diabetic cardiomyopathy in the LVEF (WT/STZ injected with adeno-associated virus carrying gfp [AAV9-gfp] vs WT/STZ AAV9-Akap1, 52.4% vs 59.6%; KO/STZ AAV9-gfp vs KO/STZ AAV9-Akap1, 42.2% vs 57.6%). CONCLUSIONS/INTERPRETATION Our study provides the first evidence that Akap1 deficiency exacerbates diabetic cardiomyopathy by impeding mitochondrial translocation of NDUFS1 to induce mitochondrial dysfunction and cardiomyocyte apoptosis. Our findings suggest that Akap1 upregulation has therapeutic potential for myocardial injury in individuals with diabetes.
Collapse
Affiliation(s)
- Bingchao Qi
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China
| | - Linjie He
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Ya Zhao
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
- Laboratory Animal Center, Fourth Military Medical University, Xi'an, China
| | - Ling Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Yuanfang He
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Jun Li
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China
| | - Bo Zhang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Qichao Huang
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
- Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Fei Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, 1 Xinsi Road, Xi'an, 710038, China.
| | - Lele Ji
- Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
31
|
Zheng Q, Wang YC, Liu QX, Dong XJ, Xie ZX, Liu XH, Gao W, Bai XJ, Li ZF. FK866 attenuates sepsis-induced acute lung injury through c-jun-N-terminal kinase (JNK)-dependent autophagy. Life Sci 2020; 250:117551. [PMID: 32179075 DOI: 10.1016/j.lfs.2020.117551] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/12/2022]
Abstract
AIMS Increasing evidence indicates that FK866, a specific noncompetitive nicotinamide phosphoribosyl transferase inhibitor, exhibits a protective effect on acute lung injury (ALI). Autophagy plays a pivotal role in sepsis-induced ALI. However, the contribution of autophagy and the underlying mechanism by which FK866-confered lung protection remains elusive. Herein, we aimed to study whether FK866 could alleviate sepsis-induced ALI via the JNK-dependent autophagy. MAIN METHODS Male C57BL/6 mice were subjected to cecal ligation and puncture (CLP) to establish the polymicrobial sepsis mice model, and treated with FK866 (10 mg/kg) at 24, 12 and 0.5 h before the CLP procedure. The lung protective effects were measured by lung histopathology, tissue edema, vascular leakage, inflammation infiltration, autophagy-related protein expression and JNK activity. A549 cells were stimulated with LPS (1000 ng/ml) to generate the ALI cell model, and pretreated with FK866 or SP600125 for 30 min to measure the autophagy-related protein expression and JNK activity. KEY FINDINGS Our results demonstrated that FK866 reduced lung injury score, tissue edema, vascular leakage, and inflammatory infiltration, and upregulated autophagy. The protective effect of autophagy conferred by FK866 on ALI was further clarified by using 3-methyladenine (3MA) and rapamycin. Additionally, the activity of JNK was suppressed by FK866, and inhibition of JNK promoted autophagy and showed a benefit effect. SIGNIFICANCE Our study indicates that FK866 protects against sepsis-induced ALI by induction of JNK-dependent autophagy. This may provide new insights into the functional mechanism of NAMPT inhibition in sepsis-induced ALI.
Collapse
Affiliation(s)
- Qiang Zheng
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Yu-Chang Wang
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Qin-Xin Liu
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Xi-Jie Dong
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Zhen-Xing Xie
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Xing-Hua Liu
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Wei Gao
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Xiang-Jun Bai
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China
| | - Zhan-Fei Li
- Trauma center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei province, China.
| |
Collapse
|
32
|
Chen Z, Ma Y, Yang Q, Hu J, Feng J, Liang W, Ding G. AKAP1 mediates high glucose-induced mitochondrial fission through the phosphorylation of Drp1 in podocytes. J Cell Physiol 2020; 235:7433-7448. [PMID: 32108342 DOI: 10.1002/jcp.29646] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/04/2020] [Indexed: 02/06/2023]
Abstract
Increasing evidence suggests that mitochondrial dysfunction plays a critical role in the development of diabetic kidney disease (DKD), however, its specific pathomechanism remains unclear. A-kinase anchoring protein (AKAP) 1 is a scaffold protein in the AKAP family that is involved in mitochondrial fission and fusion. Here, we show that rats with streptozotocin (STZ)-induced diabetes developed podocyte damage accompanied by AKAP1 overexpression and that AKAP1 closely interacted with the mitochondrial fission enzyme dynamin-related protein 1 (Drp1). At the molecular level, high glucose (HG) promoted podocyte injury and Drp1 phosphorylation at Ser637 as proven by decreased mitochondrial membrane potential, elevated reactive oxygen species generation, reduced adenosine triphosphate synthesis, and increased podocyte apoptosis. Furthermore, the AKAP1 knockdown protected HG-induced podocyte injury and suppressed HG-induced Drp1 phosphorylation at Ser637. AKAP1 overexpression aggravated HG-induced mitochondrial fragmentation and podocyte apoptosis. The coimmunoprecipitation assay showed that HG-induced Drp1 interacted with AKAP1, revealing that AKAP1 could recruit Drp1 from the cytoplasm under HG stimulation. Subsequently, we detected the effect of drp1 phosphorylation on Ser637 by transferring several different Drp1 mutants. We demonstrated that activated AKAP1 promoted Drp1 phosphorylation at Ser637, which promoted the transposition of Drp1 to the surface of the mitochondria and accounts for mitochondrial dysfunction events. These findings indicate that AKAP1 is the main pathogenic factor in the development and progression of HG-induced podocyte injury through the destruction of mitochondrial dynamic homeostasis by regulating Drp1 phosphorylation in human podocytes.
Collapse
Affiliation(s)
- Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiqiong Ma
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qian Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
33
|
A-Kinase Anchoring Protein 1: Emerging Roles in Regulating Mitochondrial Form and Function in Health and Disease. Cells 2020; 9:cells9020298. [PMID: 31991888 PMCID: PMC7072574 DOI: 10.3390/cells9020298] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 01/01/2023] Open
Abstract
Best known as the powerhouse of the cell, mitochondria have many other important functions such as buffering intracellular calcium and reactive oxygen species levels, initiating apoptosis and supporting cell proliferation and survival. Mitochondria are also dynamic organelles that are constantly undergoing fission and fusion to meet specific functional needs. These processes and functions are regulated by intracellular signaling at the mitochondria. A-kinase anchoring protein 1 (AKAP1) is a scaffold protein that recruits protein kinase A (PKA), other signaling proteins, as well as RNA to the outer mitochondrial membrane. Hence, AKAP1 can be considered a mitochondrial signaling hub. In this review, we discuss what is currently known about AKAP1's function in health and diseases. We focus on the recent literature on AKAP1's roles in metabolic homeostasis, cancer and cardiovascular and neurodegenerative diseases. In healthy tissues, AKAP1 has been shown to be important for driving mitochondrial respiration during exercise and for mitochondrial DNA replication and quality control. Several recent in vivo studies using AKAP1 knockout mice have elucidated the role of AKAP1 in supporting cardiovascular, lung and neuronal cell survival in the stressful post-ischemic environment. In addition, we discuss the unique involvement of AKAP1 in cancer tumor growth, metastasis and resistance to chemotherapy. Collectively, the data indicate that AKAP1 promotes cell survival throug regulating mitochondrial form and function. Lastly, we discuss the potential of targeting of AKAP1 for therapy of various disorders.
Collapse
|
34
|
Hegermann J, Wrede C, Fassbender S, Schliep R, Ochs M, Knudsen L, Mühlfeld C. Volume-CLEM: a method for correlative light and electron microscopy in three dimensions. Am J Physiol Lung Cell Mol Physiol 2019; 317:L778-L784. [DOI: 10.1152/ajplung.00333.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Generation of three-dimensional (3D) data sets from serial sections of tissues imaged by light microscopy (LM) allows identification of rare structures by morphology or fluorescent labeling. Here, we demonstrate a workflow for correlative LM and electron microscopy (EM) from 3D LM to 3D EM, using the same sectioned material for both methods consecutively. The new approach is easy to reproduce in routine EM laboratories and applicable to a wide range of organs and research questions.
Collapse
Affiliation(s)
- Jan Hegermann
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, Hanover, Germany
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, Hanover, Germany
| | - Susanne Fassbender
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
| | - Ronja Schliep
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
| | - Matthias Ochs
- Institute of Vegetative Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, The German Center for Lung Research (DZL), Berlin, Germany
| | - Lars Knudsen
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Research (BREATH), Member of The German Center for Lung Research (DZL), Hannover, Germany
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hanover, Germany
- Research Core Unit Electron Microscopy, Hannover Medical School, Hanover, Germany
- Biomedical Research in Endstage and Obstructive Lung Research (BREATH), Member of The German Center for Lung Research (DZL), Hannover, Germany
| |
Collapse
|
35
|
Marin W. A-kinase anchoring protein 1 (AKAP1) and its role in some cardiovascular diseases. J Mol Cell Cardiol 2019; 138:99-109. [PMID: 31783032 DOI: 10.1016/j.yjmcc.2019.11.154] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/08/2019] [Accepted: 11/22/2019] [Indexed: 01/09/2023]
Abstract
A-kinase anchoring proteins (AKAPs) play crucial roles in regulating compartmentalized multi-protein signaling networks related to PKA-mediated phosphorylation. The mitochondrial AKAP - AKAP1 proteins are enriched in heart and play cardiac protective roles. This review aims to thoroughly summarize AKAP1 variants from their sequence features to the structure-function relationships between AKAP1 and its binding partners, as well as the molecular mechanisms of AKAP1 in cardiac hypertrophy, hypoxia-induced myocardial infarction and endothelial cells dysfunction, suggesting AKAP1 as a candidate for cardiovascular therapy.
Collapse
Affiliation(s)
- Wenwen Marin
- Institute for Translational Medicine, Medical Faculty of Qingdao University, Qingdao 266021, China.
| |
Collapse
|
36
|
Dylag AM, Brookes PS, O'Reilly MA. Swapping mitochondria: a key to understanding susceptibility to neonatal chronic lung disease. Am J Physiol Lung Cell Mol Physiol 2019; 317:L737-L739. [PMID: 31596117 DOI: 10.1152/ajplung.00395.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Andrew M Dylag
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Paul S Brookes
- Department of Anesthesiology, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| | - Michael A O'Reilly
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, New York
| |
Collapse
|
37
|
The nitrated fatty acid, 10-nitrooleate inhibits the neutrophil chemotaxis via peroxisome proliferator-activated receptor gamma in CLP-induced sepsis in mice. Int Immunopharmacol 2019; 72:159-165. [DOI: 10.1016/j.intimp.2019.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 01/09/2023]
|
38
|
Protective Features of Autophagy in Pulmonary Infection and Inflammatory Diseases. Cells 2019; 8:cells8020123. [PMID: 30717487 PMCID: PMC6406971 DOI: 10.3390/cells8020123] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/29/2019] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a highly conserved catabolic process involving autolysosomal degradation of cellular components, including protein aggregates, damaged organelles (such as mitochondria, endoplasmic reticulum, and others), as well as various pathogens. Thus, the autophagy pathway represents a major adaptive response for the maintenance of cellular and tissue homeostasis in response to numerous cellular stressors. A growing body of evidence suggests that autophagy is closely associated with diverse human diseases. Specifically, acute lung injury (ALI) and inflammatory responses caused by bacterial infection or xenobiotic inhalation (e.g., chlorine and cigarette smoke) have been reported to involve a spectrum of alterations in autophagy phenotypes. The role of autophagy in pulmonary infection and inflammatory diseases could be protective or harmful dependent on the conditions. In this review, we describe recent advances regarding the protective features of autophagy in pulmonary diseases, with a focus on ALI, idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), tuberculosis, pulmonary arterial hypertension (PAH) and cystic fibrosis.
Collapse
|