1
|
Dong Z, Wang X, Wang P, Bai M, Wang T, Chu Y, Qin Y. Idiopathic Pulmonary Fibrosis Caused by Damaged Mitochondria and Imbalanced Protein Homeostasis in Alveolar Epithelial Type II Cell. Adv Biol (Weinh) 2024:e2400297. [PMID: 39390651 DOI: 10.1002/adbi.202400297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/21/2024] [Indexed: 10/12/2024]
Abstract
Alveolar epithelial Type II (ATII) cells are closely associated with early events of Idiopathic pulmonary fibrosis (IPF). Proteostasis dysfunction, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction are known causes of decreased proliferation of alveolar epithelial cells and the secretion of pro-fibrotic mediators. Here, a large body of evidence is systematized and a cascade relationship between protein homeostasis, endoplasmic reticulum stress, mitochondrial dysfunction, and fibrotropic cytokines is proposed, providing a theoretical basis for ATII cells dysfunction as a possible pathophysiological initiating event for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhaoxiong Dong
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
- Institute of Biophysics, Chinese Academy of Sciences 15 Datun Road, Chaoyang District, Beijing, 100101, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Xiaolong Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
| | - Peiwen Wang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Mingjian Bai
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100101, China
| | - Tianyu Wang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100101, China
| | - Yanhui Chu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Yan Qin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 510260, China
- Institute of Biophysics, Chinese Academy of Sciences 15 Datun Road, Chaoyang District, Beijing, 100101, China
| |
Collapse
|
2
|
Chung KP, Cheng CN, Chen YJ, Hsu CL, Huang YL, Hsieh MS, Kuo HC, Lin YT, Juan YH, Nakahira K, Chen YF, Liu WL, Ruan SY, Chien JY, Plataki M, Cloonan SM, Carmeliet P, Choi AMK, Kuo CH, Yu CJ. Alveolar epithelial cells mitigate neutrophilic inflammation in lung injury through regulating mitochondrial fatty acid oxidation. Nat Commun 2024; 15:7241. [PMID: 39174557 PMCID: PMC11341863 DOI: 10.1038/s41467-024-51683-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Type 2 alveolar epithelial (AT2) cells of the lung are fundamental in regulating alveolar inflammation in response to injury. Impaired mitochondrial long-chain fatty acid β-oxidation (mtLCFAO) in AT2 cells is assumed to aggravate alveolar inflammation in acute lung injury (ALI), yet the importance of mtLCFAO to AT2 cell function needs to be defined. Here we show that expression of carnitine palmitoyltransferase 1a (CPT1a), a mtLCFAO rate limiting enzyme, in AT2 cells is significantly decreased in acute respiratory distress syndrome (ARDS). In mice, Cpt1a deletion in AT2 cells impairs mtLCFAO without reducing ATP production and alters surfactant phospholipid abundance in the alveoli. Impairing mtLCFAO in AT2 cells via deleting either Cpt1a or Acadl (acyl-CoA dehydrogenase long chain) restricts alveolar inflammation in ALI by hindering the production of the neutrophilic chemokine CXCL2 from AT2 cells. This study thus highlights mtLCFAO as immunometabolism to injury in AT2 cells and suggests impaired mtLCFAO in AT2 cells as an anti-inflammatory response in ARDS.
Collapse
Grants
- K08 HL157728 NHLBI NIH HHS
- 109-O04, 110-O07, 110-S4872, 111-S0075, 113-S0079 National Taiwan University Hospital (NTUH)
- NTUCDP-112L7745, NTUCDP-112L7746, 110T099, NTU-NFG-110L7422 National Taiwan University (NTU)
- National Science and Technology Council (Taiwan) (MOST-108-2628-B-002-017 [K.P.C.], MOST-109-2628-B-002-044 [K.P.C.], MOST-110-2628-B-002-029 [K.P.C.], MOST-110-2628-B-002-045-MY3 [K.P.C.], MOST-111-2628-B-002-030-MY3 [K.P.C.])
- National Science and Technology Council (Taiwan), MOST 107-2314-B-002-235-MY3
- National Science and Technology Council (Taiwan), MOST 110-2314-B-002-262
- National Taiwan University School of Pharmacy Endowment Fund in support of the Platform for Clinical Mass Spectrometry and NMR Structure Elucidation
- Research funding provided by Mr. Barry Lam, the chairman of Quanta Computer Inc
Collapse
Affiliation(s)
- Kuei-Pin Chung
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chih-Ning Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Jung Chen
- Department of Laboratory Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Lin Huang
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Min-Shu Hsieh
- Department of Pathology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Han-Chun Kuo
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Ya-Ting Lin
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Hsiu Juan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kiichi Nakahira
- Department of Pharmacology, Nara Medical University, Kashihara, Nara, Japan
| | - Yen-Fu Chen
- Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Wei-Lun Liu
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
- Department of Critical Care Medicine, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei, Taiwan
| | - Sheng-Yuan Ruan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jung-Yien Chien
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Suzanne M Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Augustine M K Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Presbyterian Hospital-Weill Cornell Medical Center, New York, NY, USA
| | - Ching-Hua Kuo
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, Taiwan.
- The Metabolomics Core Laboratory, Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan.
- Department of Pharmacy, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan.
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
3
|
Zhong K, Huang Y, Chen R, Pan Q, Li J, Xi X. The protective effect of ginsenoside Rg1 against sepsis-induced lung injury through PI3K-Akt pathway: insights from molecular dynamics simulation and experimental validation. Sci Rep 2024; 14:16071. [PMID: 38992150 PMCID: PMC11239675 DOI: 10.1038/s41598-024-66908-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024] Open
Abstract
Sepsis-induced acute lung injury (SALI) poses a significant threat with high incidence and mortality rates. Ginsenoside Rg1 (GRg1), derived from Ginseng in traditional Chinese medicine, has been found to reduce inflammation and protect lung epithelial cells against tissue damage. However, the specific roles and mechanisms by which GRg1 mitigates SALI have yet to be fully elucidated. In this context, we employed a relevant SALI mouse model, alongside network pharmacology, molecular docking, and molecular dynamics simulation to pinpoint GRg1's action targets, complemented by in vitro assays to explore the underlying mechanisms. Our research shows that GRg1 alleviates CLP-induced SALI, decreasing lung tissue damage and levels of serum proinflammatory factor IL-6, TNF-α, and IL-1β, also enhancing the survival rate of CLP mice. A total of 116 common targets between GRg1 and ALI, with specific core targets including AKT1, VEGFA, SRC, IGF1, ESR1, STAT3, and ALB. Further in vitro experiments assessed GRg1's intervention effects on MLE-12 cells exposed to LPS, with qRT-PCR analysis and molecular dynamics simulations confirming AKT1 as the key target with the favorable binding activity for GRg1. Western blot results indicated that GRg1 increased the Bcl-2/Bax protein expression ratio to reduce apoptosis and decreased the high expression of cleaved caspase-3 in LPS-induced MLE-12 cells. More results showed significant increases in the phosphorylation of PI3K and AKT1. Flow cytometric analysis using PI and Annexin-V assays further verified that GRg1 decreased the apoptosis rate in LPS-stimulated MLE-12 cells (from 14.85 to 6.54%, p < 0.05). The employment of the AKT1 inhibitor LY294002 confirmed these trends, indicating that AKT1's inhibition negates GRg1's protective effects on LPS-stimulated MLE-12 cells. In conclusion, our research highlights GRg1's potential as an effective adjunct therapy for SALI, primarily by inhibiting apoptosis in alveolar epithelial cells and reducing pro-inflammatory cytokine secretion, thus significantly enhancing the survival rates of CLP mice. These beneficial effects are mediated through targeting AKT1 and activating the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Kaiqiang Zhong
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yingui Huang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Rui Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China
| | - Qiusha Pan
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jun Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, China.
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
| | - Xiaotu Xi
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, China.
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Dikalov S, Panov A, Dikalova A. Critical Role of Mitochondrial Fatty Acid Metabolism in Normal Cell Function and Pathological Conditions. Int J Mol Sci 2024; 25:6498. [PMID: 38928204 PMCID: PMC11203650 DOI: 10.3390/ijms25126498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
There is a "popular" belief that a fat-free diet is beneficial, supported by the scientific dogma indicating that high levels of fatty acids promote many pathological metabolic, cardiovascular, and neurodegenerative conditions. This dogma pressured scientists not to recognize the essential role of fatty acids in cellular metabolism and focus on the detrimental effects of fatty acids. In this work, we critically review several decades of studies and recent publications supporting the critical role of mitochondrial fatty acid metabolism in cellular homeostasis and many pathological conditions. Fatty acids are the primary fuel source and essential cell membrane building blocks from the origin of life. The essential cell membranes phospholipids were evolutionarily preserved from the earlier bacteria in human subjects. In the past century, the discovery of fatty acid metabolism was superseded by the epidemic growth of metabolic conditions and cardiovascular diseases. The association of fatty acids and pathological conditions is not due to their "harmful" effects but rather the result of impaired fatty acid metabolism and abnormal lifestyle. Mitochondrial dysfunction is linked to impaired metabolism and drives multiple pathological conditions. Despite metabolic flexibility, the loss of mitochondrial fatty acid oxidation cannot be fully compensated for by other sources of mitochondrial substrates, such as carbohydrates and amino acids, resulting in a pathogenic accumulation of long-chain fatty acids and a deficiency of medium-chain fatty acids. Despite popular belief, mitochondrial fatty acid oxidation is essential not only for energy-demanding organs such as the heart, skeletal muscle, and kidneys but also for metabolically "inactive" organs such as endothelial and epithelial cells. Recent studies indicate that the accumulation of long-chain fatty acids in specific organs and tissues support the impaired fatty acid oxidation in cell- and tissue-specific fashion. This work, therefore, provides a basis to challenge these established dogmas and articulate the need for a paradigm shift from the "pathogenic" role of fatty acids to the critical role of fatty acid oxidation. This is important to define the causative role of impaired mitochondrial fatty acid oxidation in specific pathological conditions and develop novel therapeutic approaches targeting mitochondrial fatty acid metabolism.
Collapse
Affiliation(s)
- Sergey Dikalov
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, 2220 Pierce Ave, PRB 554, Nashville, TN 37232, USA; (A.P.); (A.D.)
| | | | | |
Collapse
|
5
|
Chen H, Chen J, Feng L, Shao H, Zhou Y, Shan J, Lin L, Ye J, Wang S. Integrated network pharmacology, molecular docking, and lipidomics to reveal the regulatory effect of Qingxuan Zhike granules on lipid metabolism in lipopolysaccharide-induced acute lung injury. Biomed Chromatogr 2024; 38:e5853. [PMID: 38486466 DOI: 10.1002/bmc.5853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 05/21/2024]
Abstract
Qingxuan Zhike granules (QXZKG), a traditional Chinese patent medication, has shown therapeutic potential against acute lung injury (ALI). However, the precise mechanism underlying its lung-protective effects requires further investigation. In this study, integrated network pharmacology, molecular docking, and lipidomics were used to elucidate QXZKG's regulatory effect on lipid metabolism in lipopolysaccharide-induced ALI. Animal experiments were conducted to substantiate the efficacy of QXZKG in reducing pro-inflammatory cytokines and mitigating pulmonary pathology. Network pharmacology analysis identified 145 active compounds that directly targeted 119 primary targets of QXZKG against ALI. Gene Ontology function analysis emphasized the roles of lipid metabolism and mitogen-activated protein kinase (MAPK) cascade as crucial biological processes. The MAPK1 protein exhibited promising affinities for naringenin, luteolin, and kaempferol. Lipidomic analysis revealed that 12 lipids showed significant restoration following QXZKG treatment (p < 0.05, FC >1.2 or <0.83). Specifically, DG 38:4, DG 40:7, PC O-40:8, TG 18:1_18:3_22:6, PI 18:2_20:4, FA 16:3, FA 20:3, FA 20:4, FA 22:5, and FA 24:5 were downregulated, while Cer 18:0;2O/24:0 and SM 36:1;2O/34:5 were upregulated in the QXZKG versus model groups. This study enhances our understanding of the active compounds and targets of QXZKG, as well as the potential of lipid metabolism in the treatment of ALI.
Collapse
Affiliation(s)
- Hui Chen
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Pediatrics Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiabin Chen
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Feng
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Pediatrics Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hua Shao
- Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Yang Zhou
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Pediatrics Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Pediatrics Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lili Lin
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Pediatrics Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin Ye
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Pediatrics Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Shouchuan Wang
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Medical Metabolomics Center, Pediatrics Department, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
6
|
Xie WM, Su W, Liu XY, Zhou J, Wang M, Wang Y, Wang W, Bai X, Li Z, Li T. FTO Deficiency Alleviates LPS-induced Acute Lung Injury by TXNIP/NLRP3-mediated Alveolar Epithelial Cell Pyroptosis. Am J Respir Cell Mol Biol 2024; 70:351-363. [PMID: 38271683 DOI: 10.1165/rcmb.2023-0251oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 01/25/2024] [Indexed: 01/27/2024] Open
Abstract
N6-methyladenosine (m6A) plays a role in various diseases, but it has rarely been reported in acute lung injury (ALI). The FTO (fat mass and obesity-associated) protein can regulate mRNA metabolism by removing m6A residues. The aim of this study was to examine the role and mechanism of the m6A demethylase FTO in LPS-induced ALI. Lung epithelial FTO-knockout mice and FTO-knockdown/overexpression human alveolar epithelial (A549) cell lines were constructed to evaluate the effects of FTO on ALI. Bioinformatics analysis and a series of in vivo and in vitro assays were used to examine the mechanism of FTO regulation. Rescue assays were conducted to examine whether the impact of FTO on ALI depended on the TXNIP/NLRP3 pathway. In LPS-induced ALI, RNA m6A modification amounts were upregulated, and FTO expression was downregulated. In vivo, lung epithelial FTO knockout alleviated alveolar structure disorder, tissue edema, and pulmonary inflammation and improved the survival of ALI mice. In vitro, FTO knockdown reduced A549 cell damage and death induced by LPS, whereas FTO overexpression exacerbated cell damage and death. Mechanistically, bioinformatics analysis revealed that TXNIP was a downstream target of FTO. FTO deficiency mitigated pyroptosis in LPS-induced ALI via the TXNIP/NLRP3 pathway. Rescue assays confirmed that the impact of FTO on the TXNIP/NLRP3 pathway was significantly reversed by the TXNIP inhibitor SRI-37330. Deficiency of FTO alleviates LPS-induced ALI via TXNIP/NLRP3 pathway-mediated alveolar epithelial cell pyroptosis, which might be a novel therapeutic strategy for combating ALI.
Collapse
Affiliation(s)
- Wei-Ming Xie
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, and
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Wei Su
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, and
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Xin-Yu Liu
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, and
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Junhao Zhou
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Wuhan, China
| | - Min Wang
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Yuchang Wang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, and
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Wei Wang
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, and
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Xiangjun Bai
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, and
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Zhanfei Li
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, and
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| | - Tianyu Li
- Division of Trauma Surgery, Emergency Surgery & Surgical Critical, Tongji Trauma Center, and
- Department of Emergency and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; and
| |
Collapse
|
7
|
Polverino F, Mora A. Alveolar Epithelial Cell Dysfunction in Idiopathic Pulmonary Fibrosis Linked to Lipid Alterations: Therapeutic Implications. Am J Respir Cell Mol Biol 2024; 70:233-234. [PMID: 38271680 PMCID: PMC11478126 DOI: 10.1165/rcmb.2023-0432ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/23/2024] [Indexed: 01/27/2024] Open
Affiliation(s)
| | - Ana Mora
- Division of Pulmonary, Critical Care, and Sleep Medicine Ohio State University Columbus, Ohio
| |
Collapse
|
8
|
Lu X, Li G, Liu Y, Luo G, Ding S, Zhang T, Li N, Geng Q. The role of fatty acid metabolism in acute lung injury: a special focus on immunometabolism. Cell Mol Life Sci 2024; 81:120. [PMID: 38456906 PMCID: PMC10923746 DOI: 10.1007/s00018-024-05131-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/06/2024] [Accepted: 01/17/2024] [Indexed: 03/09/2024]
Abstract
Reputable evidence from multiple studies suggests that excessive and uncontrolled inflammation plays an indispensable role in mediating, amplifying, and protracting acute lung injury (ALI). Traditionally, immunity and energy metabolism are regarded as separate functions regulated by distinct mechanisms, but recently, more and more evidence show that immunity and energy metabolism exhibit a strong interaction which has given rise to an emerging field of immunometabolism. Mammalian lungs are organs with active fatty acid metabolism, however, during ALI, inflammation and oxidative stress lead to a series metabolic reprogramming such as impaired fatty acid oxidation, increased expression of proteins involved in fatty acid uptake and transport, enhanced synthesis of fatty acids, and accumulation of lipid droplets. In addition, obesity represents a significant risk factor for ALI/ARDS. Thus, we have further elucidated the mechanisms of obesity exacerbating ALI from the perspective of fatty acid metabolism. To sum up, this paper presents a systematical review of the relationship between extensive fatty acid metabolic pathways and acute lung injury and summarizes recent advances in understanding the involvement of fatty acid metabolism-related pathways in ALI. We hold an optimistic believe that targeting fatty acid metabolism pathway is a promising lung protection strategy, but the specific regulatory mechanisms are way too complex, necessitating further extensive and in-depth investigations in future studies.
Collapse
Affiliation(s)
- Xiao Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Guoqing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Tianyu Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
9
|
Wang GY, Xu X, Xiong DY, Deng L, Liu W, Huang XT. CPT1A as a potential therapeutic target for lipopolysaccharide-induced acute lung injury in mice. Sci Rep 2024; 14:1600. [PMID: 38238472 PMCID: PMC10796431 DOI: 10.1038/s41598-024-52042-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024] Open
Abstract
Acute lung injury (ALI) remains a high mortality rate with dramatic lung inflammation and alveolar epithelial cell death. Although fatty acid β-oxidation (FAO) impairment has been implicated in the pathogenesis of ALI, whether Carnitine palmitoyltransferase 1A (CPT1A), the rate-limiting enzyme for FAO, plays roles in lipopolysaccharide (LPS)-induced ALI remains unclear. Accordingly, we focused on exploring the effect of CPT1A in the context of ALI and the underlying mechanisms. We found that overexpression of CPT1A (AAV-CPT1A) effectively alleviated lung injury by reduction of lung wet-to-dry ratio, inflammatory cell infiltration, and protein levels in the BALF of ALI mice. Meanwhile, AAV-CPT1A significantly lessened histopathological changes and several cytokines' secretions. In contrast, blocking CPT1A with etomoxir augmented inflammatory responses and lung injury in ALI mice. Furthermore, we found that overexpression of CPT1A with lentivirus reduced the apoptosis rates of alveolar epithelial cells and the expression of apoptosis-related proteins induced by LPS in MLE12 cells, while etomoxir increased the apoptosis of MLE12 cells. Overexpression of CPT1A prevented the drop in bioenergetics, palmitate oxidation, and ATP levels. In conclusion, the results rendered CPT1A worthy of further development into a pharmaceutical drug for the treatment of ALI.
Collapse
Affiliation(s)
- Gui-Yun Wang
- Shandong Xiehe University, Jinan, 250109, Shandong, China
| | - Xia Xu
- Shandong Xiehe University, Jinan, 250109, Shandong, China
| | - Da-Yan Xiong
- Xiangya School of Nursing, Central South University, Changsha, 410013, Hunan, China
| | - Lang Deng
- Xiangya School of Nursing, Central South University, Changsha, 410013, Hunan, China
| | - Wei Liu
- Xiangya School of Nursing, Central South University, Changsha, 410013, Hunan, China
| | - Xiao-Ting Huang
- Xiangya School of Nursing, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
10
|
Miguel V, Rey-Serra C, Tituaña J, Sirera B, Alcalde-Estévez E, Herrero JI, Ranz I, Fernández L, Castillo C, Sevilla L, Nagai J, Reimer KC, Jansen J, Kramann R, Costa IG, Castro A, Sancho D, Rodríguez González-Moro JM, Lamas S. Enhanced fatty acid oxidation through metformin and baicalin as therapy for COVID-19 and associated inflammatory states in lung and kidney. Redox Biol 2023; 68:102957. [PMID: 37977043 PMCID: PMC10682832 DOI: 10.1016/j.redox.2023.102957] [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: 09/25/2023] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023] Open
Abstract
Progressive respiratory failure is the primary cause of death in the coronavirus disease 2019 (COVID-19) pandemic. It is the final outcome of the acute respiratory distress syndrome (ARDS), characterized by an initial exacerbated inflammatory response, metabolic derangement and ultimate tissue scarring. A positive balance of cellular energy may result crucial for the recovery of clinical COVID-19. Hence, we asked if two key pathways involved in cellular energy generation, AMP-activated protein kinase (AMPK)/acetyl-CoA carboxylase (ACC) signaling and fatty acid oxidation (FAO) could be beneficial. We tested the drugs metformin (AMPK activator) and baicalin (CPT1A activator) in different experimental models mimicking COVID-19 associated inflammation in lung and kidney. We also studied two different cohorts of COVID-19 patients that had been previously treated with metformin. These drugs ameliorated lung damage in an ARDS animal model, while activation of AMPK/ACC signaling increased mitochondrial function and decreased TGF-β-induced fibrosis, apoptosis and inflammation markers in lung epithelial cells. Similar results were observed with two indole derivatives, IND6 and IND8 with AMPK activating capacity. Consistently, a reduced time of hospitalization and need of intensive care was observed in COVID-19 patients previously exposed to metformin. Baicalin also mitigated the activation of pro-inflammatory bone marrow-derived macrophages (BMDMs) and reduced kidney fibrosis in two animal models of kidney injury, another key target of COVID-19. In human epithelial lung and kidney cells, both drugs improved mitochondrial function and prevented TGF-β-induced renal epithelial cell dedifferentiation. Our results support that favoring cellular energy production through enhanced FAO may prove useful in the prevention of COVID-19-induced lung and renal damage.
Collapse
Affiliation(s)
- Verónica Miguel
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029, Madrid, Spain.
| | - Carlos Rey-Serra
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Jessica Tituaña
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Belén Sirera
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Elena Alcalde-Estévez
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - J Ignacio Herrero
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Irene Ranz
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Laura Fernández
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain
| | - Carolina Castillo
- Department of Pathology. University Hospital "Príncipe de Asturias", Alcalá de Henares, Madrid, Spain
| | - Lucía Sevilla
- Department of Pneumology, University Hospital "Principe de Asturias", Alcala de Henares, Madrid, Spain
| | - James Nagai
- Institute for Computational Genomics, RWTH Aachen University Hospital, Aachen, Germany; Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Katharina C Reimer
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany; Institute for Biomedical Technologies, Department of Cell Biology, RWTH Aachen University, Aachen, Germany
| | - Jitske Jansen
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany; Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rafael Kramann
- Department of Medicine 2, Nephrology, Rheumatology and Immunology, RWTH Aachen University, Medical Faculty, Aachen, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, RWTH Aachen University Hospital, Aachen, Germany; Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Ana Castro
- Instituto de Química Medica (IQM-CSIC), Juan de la Cierva 3, 28006, Madrid, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029, Madrid, Spain
| | | | - Santiago Lamas
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa" (CBMSO) (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
11
|
Wu D, He H, Chen J, Yao S, Xie H, Jiang W, Lv X, Gao W, Meng L, Yao X. L-carnitine reduces acute lung injury via mitochondria modulation and inflammation control in pulmonary macrophages. Braz J Med Biol Res 2023; 56:e12830. [PMID: 37878885 PMCID: PMC10591484 DOI: 10.1590/1414-431x2023e12830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/11/2023] [Indexed: 10/27/2023] Open
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a critical respiratory syndrome with limited effective interventions. Lung macrophages play a critical role in the pathogenesis of abnormal inflammatory response in the syndrome. Recently, impaired fatty acid oxidation (FAO), one of the key lipid metabolic signalings, was found to participate in the onset and development of various lung diseases, including ALI/ARDS. Lipid/fatty acid contents within mouse lungs were quantified using the Oil Red O staining. The protective effect of FAO activator L-carnitine (Lca, 50, 500, or 5 mg/mL) was evaluated by cell counting kit 8 (CCK-8) assay, real-time quantitative PCR (qPCR), ELISA, immunoblotting, fluorescence imaging, and fluorescence plate reader detection in lipopolysaccharide (LPS) (100 ng/mL)-stimulated THP-1-derived macrophages. The in vivo efficacy of Lca (300 mg/kg) was determined in a 10 mg/kg LPS-induced ALI mouse model. We found for the first time that lipid accumulation in pulmonary macrophages was significantly increased in a classical ALI murine model, which indicated disrupted FAO induced by LPS. Lca showed potent anti-inflammatory and antioxidative effects on THP-1 derived macrophages upon LPS stimulation. Mechanistically, Lca was able to maintain FAO, mitochondrial activity, and ameliorate mitochondrial dynamics. In the LPS-induced ALI mouse model, we further discovered that Lca inhibited neutrophilic inflammation and decreased diffuse damage, which might be due to the preservation of mitochondrial homeostasis. These results broadened our understanding of ALI/ARDS pathogenesis and provided a promising drug candidate for this syndrome.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou, Nanjing, China
| | - Haiyan He
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nantong University, North Haierxiang, Nantong, China
| | - Jinliang Chen
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nantong University, North Haierxiang, Nantong, China
| | - Sumei Yao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nantong University, North Haierxiang, Nantong, China
| | - Haiqin Xie
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nantong University, North Haierxiang, Nantong, China
| | - Wenyan Jiang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nantong University, North Haierxiang, Nantong, China
| | - Xuedong Lv
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nantong University, North Haierxiang, Nantong, China
| | - Wei Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Jimo, Shanghai, China
| | - Linlin Meng
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Jimo, Shanghai, China
| | - Xin Yao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou, Nanjing, China
| |
Collapse
|
12
|
Fan LC, McConn K, Plataki M, Kenny S, Williams NC, Kim K, Quirke JA, Chen Y, Sauler M, Möbius ME, Chung KP, Area Gomez E, Choi AM, Xu JF, Cloonan SM. Alveolar type II epithelial cell FASN maintains lipid homeostasis in experimental COPD. JCI Insight 2023; 8:e163403. [PMID: 37606038 PMCID: PMC10543729 DOI: 10.1172/jci.insight.163403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/10/2023] [Indexed: 08/23/2023] Open
Abstract
Alveolar epithelial type II (AEC2) cells strictly regulate lipid metabolism to maintain surfactant synthesis. Loss of AEC2 cell function and surfactant production are implicated in the pathogenesis of the smoking-related lung disease chronic obstructive pulmonary disease (COPD). Whether smoking alters lipid synthesis in AEC2 cells and whether altering lipid metabolism in AEC2 cells contributes to COPD development are unclear. In this study, high-throughput lipidomic analysis revealed increased lipid biosynthesis in AEC2 cells isolated from mice chronically exposed to cigarette smoke (CS). Mice with a targeted deletion of the de novo lipogenesis enzyme, fatty acid synthase (FASN), in AEC2 cells (FasniΔAEC2) exposed to CS exhibited higher bronchoalveolar lavage fluid (BALF) neutrophils, higher BALF protein, and more severe airspace enlargement. FasniΔAEC2 mice exposed to CS had lower levels of key surfactant phospholipids but higher levels of BALF ether phospholipids, sphingomyelins, and polyunsaturated fatty acid-containing phospholipids, as well as increased BALF surface tension. FasniΔAEC2 mice exposed to CS also had higher levels of protective ferroptosis markers in the lung. These data suggest that AEC2 cell FASN modulates the response of the lung to smoke by regulating the composition of the surfactant phospholipidome.
Collapse
Affiliation(s)
- Li-Chao Fan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Keith McConn
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Maria Plataki
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Sarah Kenny
- School of Medicine, Trinity Biomedical Sciences Institute, and
| | | | - Kihwan Kim
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | | | - Yan Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Maor Sauler
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | | | - Kuei-Pin Chung
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Laboratory Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Estela Area Gomez
- Division of Neuromuscular Medicine, Department of Neurology, Columbia University Irving Medical Center, Neurological Institute, New York, New York, USA
- Center for Biological Research “Margarita Salas”, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Augustine M.K. Choi
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- NewYork-Presbyterian Hospital, Weill Cornell Medicine, New York, New York, USA
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Suzanne M. Cloonan
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- School of Medicine, Trinity Biomedical Sciences Institute, and
| |
Collapse
|
13
|
Zhang L, Wu L, Zhu X, Mei J, Chen Y. Paeonol represses A549 cell glycolytic reprogramming and proliferation by decreasing m6A modification of Acyl-CoA dehydrogenase. CHINESE J PHYSIOL 2023; 66:248-256. [PMID: 37635484 DOI: 10.4103/cjop.cjop-d-22-00166] [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: 08/29/2023] Open
Abstract
Aberrant glycolytic reprogramming is involved in lung cancer progression by promoting the proliferation of non-small cell lung cancer cells. Paeonol, as a traditional Chinese medicine, plays a critical role in multiple cancer cell proliferation and inflammation. Acyl-CoA dehydrogenase (ACADM) is involved in the development of metabolic diseases. N6-methyladenosine (m6A) modification is important for the regulation of messenger RNA stability, splicing, and translation. Here, we investigated whether paeonol regulates the proliferation and glycolytic reprogramming via ACADM with m6A modification in A549 cells (human non-small cell lung cancer cells). Cell counting kit 8, 5-Bromo-2-deoxyuridine, 5-ethynyl-2'-deoxyuridine (EdU) incorporation, flow cytometry analysis, western blotting and seahorse XFe24 extracellular flux analyzer assays showed that paeonol had a significant inhibitory effect against A549 cell proliferation and glycolysis. Mechanistically, ACADM was a functional target of paeonol. We also showed that the m6A reader YTH domain containing 1 plays an important role in m6A-modified ACADM expression, which is negatively regulated by paeonol, and is involved in A549 cell proliferation and glycolytic reprogramming. These results indicated the central function of paeonol in regulating A549 cell glycolytic reprogramming and proliferation via m6A modification of ACADM.
Collapse
Affiliation(s)
- Lixin Zhang
- Central Laboratory of Harbin Medical University, Daqing, China; Department of Immunology, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Lihua Wu
- Department of Geriatrics, Daqing Oilfield General Hospital, Daqing, China
| | - Xiangrui Zhu
- Department of Immunology, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Jian Mei
- Department of Immunology, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| | - Yingli Chen
- Central Laboratory of Harbin Medical University, Daqing, China; Department of Immunology, College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, China
| |
Collapse
|
14
|
Suber TL, Wendell SG, Mullett SJ, Zuchelkowski B, Bain W, Kitsios GD, McVerry BJ, Ray P, Ray A, Mallampalli RK, Zhang Y, Shah F, Nouraie SM, Lee JS. Serum metabolomic signatures of fatty acid oxidation defects differentiate host-response subphenotypes of acute respiratory distress syndrome. Respir Res 2023; 24:136. [PMID: 37210531 PMCID: PMC10199668 DOI: 10.1186/s12931-023-02447-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 05/09/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Fatty acid oxidation (FAO) defects have been implicated in experimental models of acute lung injury and associated with poor outcomes in critical illness. In this study, we examined acylcarnitine profiles and 3-methylhistidine as markers of FAO defects and skeletal muscle catabolism, respectively, in patients with acute respiratory failure. We determined whether these metabolites were associated with host-response ARDS subphenotypes, inflammatory biomarkers, and clinical outcomes in acute respiratory failure. METHODS In a nested case-control cohort study, we performed targeted analysis of serum metabolites of patients intubated for airway protection (airway controls), Class 1 (hypoinflammatory), and Class 2 (hyperinflammatory) ARDS patients (N = 50 per group) during early initiation of mechanical ventilation. Relative amounts were quantified by liquid chromatography high resolution mass spectrometry using isotope-labeled standards and analyzed with plasma biomarkers and clinical data. RESULTS Of the acylcarnitines analyzed, octanoylcarnitine levels were twofold increased in Class 2 ARDS relative to Class 1 ARDS or airway controls (P = 0.0004 and < 0.0001, respectively) and was positively associated with Class 2 by quantile g-computation analysis (P = 0.004). In addition, acetylcarnitine and 3-methylhistidine were increased in Class 2 relative to Class 1 and positively correlated with inflammatory biomarkers. In all patients within the study with acute respiratory failure, increased 3-methylhistidine was observed in non-survivors at 30 days (P = 0.0018), while octanoylcarnitine was increased in patients requiring vasopressor support but not in non-survivors (P = 0.0001 and P = 0.28, respectively). CONCLUSIONS This study demonstrates that increased levels of acetylcarnitine, octanoylcarnitine, and 3-methylhistidine distinguish Class 2 from Class 1 ARDS patients and airway controls. Octanoylcarnitine and 3-methylhistidine were associated with poor outcomes in patients with acute respiratory failure across the cohort independent of etiology or host-response subphenotype. These findings suggest a role for serum metabolites as biomarkers in ARDS and poor outcomes in critically ill patients early in the clinical course.
Collapse
Affiliation(s)
- Tomeka L Suber
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA.
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Stacy G Wendell
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steven J Mullett
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Benjamin Zuchelkowski
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - William Bain
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Georgios D Kitsios
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bryan J McVerry
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Prabir Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anuradha Ray
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Rama K Mallampalli
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - Faraaz Shah
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
- Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Seyed Mehdi Nouraie
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Montefiore Hospital, University of Pittsburgh School of Medicine, NW 628, 3459 Fifth Avenue, Pittsburgh, PA, 15213, USA
| | - Janet S Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University at St. Louis, St. Louis, MO, USA
| |
Collapse
|
15
|
Li N, Liu B, Xiong R, Li G, Wang B, Geng Q. HDAC3 deficiency protects against acute lung injury by maintaining epithelial barrier integrity through preserving mitochondrial quality control. Redox Biol 2023; 63:102746. [PMID: 37244125 DOI: 10.1016/j.redox.2023.102746] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/12/2023] [Indexed: 05/29/2023] Open
Abstract
Sepsis is one common cause of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), which is closely associated with high mortality in intensive care units (ICU). Histone deacetylase 3 (HDAC3) serves as an important epigenetic modifying enzyme which could affect chromatin structure and transcriptional regulation. Here, we explored the effects of HDAC3 in type II alveolar epithelial cells (AT2) on lipopolysaccharide (LPS)-induced ALI and shed light on potential molecular mechanisms. We generated ALI mouse model with HDAC3 conditional knockout mice (Sftpc-cre; Hdac3f/f) in AT2 and the roles of HDAC3 in ALI and epithelial barrier integrity were investigated in LPS-treated AT2. The levels of HDAC3 were significantly upregulated in lung tissues from mice with sepsis and in LPS-treated AT2. HDAC3 deficiency in AT2 not only decreased inflammation, apoptosis, and oxidative stress, but also maintained epithelial barrier integrity. Meanwhile, HDAC3 deficiency in LPS-treated AT2 preserved mitochondrial quality control (MQC), evidenced by the shift of mitochondria from fission into fusion, decreased mitophagy, and improved fatty acid oxidation (FAO). Mechanically, HDAC3 promoted the transcription of Rho-associated protein kinase 1 (ROCK1) in AT2. In the context of LPS stimulation, the upregulated ROCK1 elicited by HDAC3 could be phosphorylated by Rho-associated (RhoA), thus disturbing MQC and triggering ALI. Furthermore, we found that forkhead box O1 (FOXO1) was one of transcription factors of ROCK1. HDAC3 directly decreased the acetylation of FOXO1 and promoted its nuclear translocation in LPS-treated AT2. Finally, HDAC3 inhibitor RGFP966 alleviated epithelial damage and improved MQC in LPS-treated AT2. Altogether, HDAC3 deficiency in AT2 alleviated sepsis-induced ALI by preserving mitochondrial quality control via FOXO1-ROCK1 axis, which provided a potential strategy for the treatment of sepsis and ALI.
Collapse
Affiliation(s)
- Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bohao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China; Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Rui Xiong
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guorui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bo Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
16
|
Faherty L, Kenny S, Cloonan SM. Iron and mitochondria in the susceptibility, pathogenesis and progression of COPD. Clin Sci (Lond) 2023; 137:219-237. [PMID: 36729089 DOI: 10.1042/cs20210504] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 02/03/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a debilitating lung disease characterised by airflow limitation, chronic bronchitis, emphysema and airway remodelling. Cigarette smoke is considered the primary risk factor for the development of COPD; however, genetic factors, host responses and infection also play an important role. Accumulating evidence highlights a role for iron dyshomeostasis and cellular iron accumulation in the lung as a key contributing factor in the development and pathogenesis of COPD. Recent studies have also shown that mitochondria, the central players in cellular iron utilisation, are dysfunctional in respiratory cells in individuals with COPD, with alterations in mitochondrial bioenergetics and dynamics driving disease progression. Understanding the molecular mechanisms underlying the dysfunction of mitochondria and cellular iron metabolism in the lung may unveil potential novel investigational avenues and therapeutic targets to aid in the treatment of COPD.
Collapse
Affiliation(s)
- Lynne Faherty
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Sarah Kenny
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Suzanne M Cloonan
- School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, New York, NY, U.S.A
| |
Collapse
|
17
|
Crotta S, Villa M, Major J, Finsterbusch K, Llorian M, Carmeliet P, Buescher J, Wack A. Repair of airway epithelia requires metabolic rewiring towards fatty acid oxidation. Nat Commun 2023; 14:721. [PMID: 36781848 PMCID: PMC9925445 DOI: 10.1038/s41467-023-36352-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 01/27/2023] [Indexed: 02/15/2023] Open
Abstract
Epithelial tissues provide front-line barriers shielding the organism from invading pathogens and harmful substances. In the airway epithelium, the combined action of multiciliated and secretory cells sustains the mucociliary escalator required for clearance of microbes and particles from the airways. Defects in components of mucociliary clearance or barrier integrity are associated with recurring infections and chronic inflammation. The timely and balanced differentiation of basal cells into mature epithelial cell subsets is therefore tightly controlled. While different growth factors regulating progenitor cell proliferation have been described, little is known about the role of metabolism in these regenerative processes. Here we show that basal cell differentiation correlates with a shift in cellular metabolism from glycolysis to fatty acid oxidation (FAO). We demonstrate both in vitro and in vivo that pharmacological and genetic impairment of FAO blocks the development of fully differentiated airway epithelial cells, compromising the repair of airway epithelia. Mechanistically, FAO links to the hexosamine biosynthesis pathway to support protein glycosylation in airway epithelial cells. Our findings unveil the metabolic network underpinning the differentiation of airway epithelia and identify novel targets for intervention to promote lung repair.
Collapse
Affiliation(s)
- Stefania Crotta
- Immunoregulation Laboratory, Francis Crick Institute, London, UK.
| | - Matteo Villa
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Jack Major
- Immunoregulation Laboratory, Francis Crick Institute, London, UK
| | | | | | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Center for Cancer Biology, VIB, and Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Biotechnology (BTC), Khalifa University of Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Joerg Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Andreas Wack
- Immunoregulation Laboratory, Francis Crick Institute, London, UK.
| |
Collapse
|
18
|
Alkhayyat SS, Al-kuraishy HM, Al-Gareeb AI, El-Bouseary MM, AboKamer AM, Batiha GES, Simal-Gandara J. Fenofibrate for COVID-19 and related complications as an approach to improve treatment outcomes: the missed key for Holy Grail. Inflamm Res 2022; 71:1159-1167. [PMID: 35941297 PMCID: PMC9360649 DOI: 10.1007/s00011-022-01615-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/05/2022] [Accepted: 07/17/2022] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Fenofibrate is an agonist of peroxisome proliferator activated receptor alpha (PPAR-α), that possesses anti-inflammatory, antioxidant, and anti-thrombotic properties. Fenofibrate is effective against a variety of viral infections and different inflammatory disorders. Therefore, the aim of critical review was to overview the potential role of fenofibrate in the pathogenesis of SARS-CoV-2 and related complications. RESULTS By destabilizing SARS-CoV-2 spike protein and preventing it from binding angiotensin-converting enzyme 2 (ACE2), a receptor for SARS-CoV-2 entry, fenofibrate can reduce SARS-CoV-2 entry in human cells Fenofibrate also suppresses inflammatory signaling pathways, which decreases SARS-CoV-2 infection-related inflammatory alterations. In conclusion, fenofibrate anti-inflammatory, antioxidant, and antithrombotic capabilities may help to minimize the inflammatory and thrombotic consequences associated with SARSCoV-2 infection. Through attenuating the interaction between SARS-CoV-2 and ACE2, fenofibrate can directly reduce the risk of SARS-CoV-2 infection. CONCLUSIONS As a result, fenofibrate could be a potential treatment approach for COVID-19 control.
Collapse
Affiliation(s)
- Shadi Salem Alkhayyat
- Department of Internal Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
| | - Hayder M. Al-kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
| | - Ali I. Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
| | - Maisra M. El-Bouseary
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amal M. AboKamer
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511 Al Beheira Egypt
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty Science, Universidade de Vigo, 32004 Ourense, Spain
| |
Collapse
|
19
|
Hu Q, Zhang S, Yang Y, Yao JQ, Tang WF, Lyon CJ, Hu TY, Wan MH. Extracellular vesicles in the pathogenesis and treatment of acute lung injury. Mil Med Res 2022; 9:61. [PMID: 36316787 PMCID: PMC9623953 DOI: 10.1186/s40779-022-00417-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common life-threatening lung diseases associated with acute and severe inflammation. Both have high mortality rates, and despite decades of research on clinical ALI/ARDS, there are no effective therapeutic strategies. Disruption of alveolar-capillary barrier integrity or activation of inflammatory responses leads to lung inflammation and injury. Recently, studies on the role of extracellular vesicles (EVs) in regulating normal and pathophysiologic cell activities, including inflammation and injury responses, have attracted attention. Injured and dysfunctional cells often secrete EVs into serum or bronchoalveolar lavage fluid with altered cargoes, which can be used to diagnose and predict the development of ALI/ARDS. EVs secreted by mesenchymal stem cells can also attenuate inflammatory reactions associated with cell dysfunction and injury to preserve or restore cell function, and thereby promote cell proliferation and tissue regeneration. This review focuses on the roles of EVs in the pathogenesis of pulmonary inflammation, particularly ALI/ARDS.
Collapse
Affiliation(s)
- Qian Hu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Shu Zhang
- Department of Emergency Medicine, Emergency Medical Laboratory, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yue Yang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Jia-Qi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Wen-Fu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Christopher J Lyon
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA.,Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA
| | - Tony Ye Hu
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA. .,Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA.
| | - Mei-Hua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China. .,West China Hospital (Airport) of Sichuan University, Chengdu, 610299, China.
| |
Collapse
|
20
|
Sanches Santos Rizzo Zuttion M, Moore SKL, Chen P, Beppu AK, Hook JL. New Insights into the Alveolar Epithelium as a Driver of Acute Respiratory Distress Syndrome. Biomolecules 2022; 12:biom12091273. [PMID: 36139112 PMCID: PMC9496395 DOI: 10.3390/biom12091273] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
The alveolar epithelium serves as a barrier between the body and the external environment. To maintain efficient gas exchange, the alveolar epithelium has evolved to withstand and rapidly respond to an assortment of inhaled, injury-inducing stimuli. However, alveolar damage can lead to loss of alveolar fluid barrier function and exuberant, non-resolving inflammation that manifests clinically as acute respiratory distress syndrome (ARDS). This review discusses recent discoveries related to mechanisms of alveolar homeostasis, injury, repair, and regeneration, with a contemporary emphasis on virus-induced lung injury. In addition, we address new insights into how the alveolar epithelium coordinates injury-induced lung inflammation and review maladaptive lung responses to alveolar damage that drive ARDS and pathologic lung remodeling.
Collapse
Affiliation(s)
- Marilia Sanches Santos Rizzo Zuttion
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sarah Kathryn Littlehale Moore
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peter Chen
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew Kota Beppu
- Women’s Guild Lung Institute, Division of Pulmonary and Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jaime Lynn Hook
- Lung Imaging Laboratory, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Correspondence:
| |
Collapse
|
21
|
Batra R, Whalen W, Alvarez-Mulett S, Gomez-Escobar LG, Hoffman KL, Simmons W, Harrington J, Chetnik K, Buyukozkan M, Benedetti E, Choi ME, Suhre K, Schenck E, Choi AMK, Schmidt F, Cho SJ, Krumsiek J. Multi-omic comparative analysis of COVID-19 and bacterial sepsis-induced ARDS. PLoS Pathog 2022; 18:e1010819. [PMID: 36121875 PMCID: PMC9484674 DOI: 10.1371/journal.ppat.1010819] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/19/2022] [Indexed: 12/06/2022] Open
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS), a life-threatening condition characterized by hypoxemia and poor lung compliance, is associated with high mortality. ARDS induced by COVID-19 has similar clinical presentations and pathological manifestations as non-COVID-19 ARDS. However, COVID-19 ARDS is associated with a more protracted inflammatory respiratory failure compared to traditional ARDS. Therefore, a comprehensive molecular comparison of ARDS of different etiologies groups may pave the way for more specific clinical interventions. METHODS AND FINDINGS In this study, we compared COVID-19 ARDS (n = 43) and bacterial sepsis-induced (non-COVID-19) ARDS (n = 24) using multi-omic plasma profiles covering 663 metabolites, 1,051 lipids, and 266 proteins. To address both between- and within- ARDS group variabilities we followed two approaches. First, we identified 706 molecules differently abundant between the two ARDS etiologies, revealing more than 40 biological processes differently regulated between the two groups. From these processes, we assembled a cascade of therapeutically relevant pathways downstream of sphingosine metabolism. The analysis suggests a possible overactivation of arginine metabolism involved in long-term sequelae of ARDS and highlights the potential of JAK inhibitors to improve outcomes in bacterial sepsis-induced ARDS. The second part of our study involved the comparison of the two ARDS groups with respect to clinical manifestations. Using a data-driven multi-omic network, we identified signatures of acute kidney injury (AKI) and thrombocytosis within each ARDS group. The AKI-associated network implicated mitochondrial dysregulation which might lead to post-ARDS renal-sequalae. The thrombocytosis-associated network hinted at a synergy between prothrombotic processes, namely IL-17, MAPK, TNF signaling pathways, and cell adhesion molecules. Thus, we speculate that combination therapy targeting two or more of these processes may ameliorate thrombocytosis-mediated hypercoagulation. CONCLUSION We present a first comprehensive molecular characterization of differences between two ARDS etiologies-COVID-19 and bacterial sepsis. Further investigation into the identified pathways will lead to a better understanding of the pathophysiological processes, potentially enabling novel therapeutic interventions.
Collapse
Affiliation(s)
- Richa Batra
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - William Whalen
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Sergio Alvarez-Mulett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Luis G. Gomez-Escobar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Katherine L. Hoffman
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, New York, United States of America
| | - Will Simmons
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, New York, United States of America
| | - John Harrington
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Kelsey Chetnik
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Mustafa Buyukozkan
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Elisa Benedetti
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Mary E. Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, New York, United States of America
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine–Qatar, Qatar Foundation, Doha, Qatar
| | - Edward Schenck
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Augustine M. K. Choi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medicine–Qatar, Qatar Foundation, Doha, Qatar
| | - Soo Jung Cho
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, United States of America
| |
Collapse
|
22
|
Batra R, Whalen W, Alvarez-Mulett S, Gómez-Escobar LG, Hoffman KL, Simmons W, Harrington J, Chetnik K, Buyukozkan M, Benedetti E, Choi ME, Suhre K, Schenck E, Choi AMK, Schmidt F, Cho SJ, Krumsiek J. Multi-omic comparative analysis of COVID-19 and bacterial sepsis-induced ARDS. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.05.16.22274587. [PMID: 35982655 PMCID: PMC9387161 DOI: 10.1101/2022.05.16.22274587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Acute respiratory distress syndrome (ARDS), a life-threatening condition characterized by hypoxemia and poor lung compliance, is associated with high mortality. ARDS induced by COVID-19 has similar clinical presentations and pathological manifestations as non-COVID-19 ARDS. However, COVID-19 ARDS is associated with a more protracted inflammatory respiratory failure compared to traditional ARDS. Therefore, a comprehensive molecular comparison of ARDS of different etiologies groups may pave the way for more specific clinical interventions. Methods and Findings In this study, we compared COVID-19 ARDS (n=43) and bacterial sepsis-induced (non-COVID-19) ARDS (n=24) using multi-omic plasma profiles covering 663 metabolites, 1,051 lipids, and 266 proteins. To address both between- and within-ARDS group variabilities we followed two approaches. First, we identified 706 molecules differently abundant between the two ARDS etiologies, revealing more than 40 biological processes differently regulated between the two groups. From these processes, we assembled a cascade of therapeutically relevant pathways downstream of sphingosine metabolism. The analysis suggests a possible overactivation of arginine metabolism involved in long-term sequelae of ARDS and highlights the potential of JAK inhibitors to improve outcomes in bacterial sepsis-induced ARDS. The second part of our study involved the comparison of the two ARDS groups with respect to clinical manifestations. Using a data-driven multi-omic network, we identified signatures of acute kidney injury (AKI) and thrombocytosis within each ARDS group. The AKI-associated network implicated mitochondrial dysregulation which might lead to post-ARDS renal-sequalae. The thrombocytosis-associated network hinted at a synergy between prothrombotic processes, namely IL-17, MAPK, TNF signaling pathways, and cell adhesion molecules. Thus, we speculate that combination therapy targeting two or more of these processes may ameliorate thrombocytosis-mediated hypercoagulation. Conclusion We present a first comprehensive molecular characterization of differences between two ARDS etiologies - COVID-19 and bacterial sepsis. Further investigation into the identified pathways will lead to a better understanding of the pathophysiological processes, potentially enabling novel therapeutic interventions.
Collapse
Affiliation(s)
- Richa Batra
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - William Whalen
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sergio Alvarez-Mulett
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Luis G Gómez-Escobar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Katherine L Hoffman
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, NY, USA
| | - Will Simmons
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, NY, USA
| | - John Harrington
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Kelsey Chetnik
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mustafa Buyukozkan
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Elisa Benedetti
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Mary E Choi
- Division of Nephrology and Hypertension, Joan and Sanford I. Weill Department of Medicine, New York, NY, USA
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine - Qatar, Qatar Foundation, Doha, Qatar
| | - Edward Schenck
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Augustine M K Choi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Frank Schmidt
- Proteomics Core, Weill Cornell Medicine - Qatar, Qatar Foundation, Doha, Qatar
| | - Soo Jung Cho
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jan Krumsiek
- Department of Physiology and Biophysics, Institute for Computational Biomedicine, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| |
Collapse
|
23
|
Nadamuni M, Venable AH, Huen SC. When a calorie isn't just a calorie: a revised look at nutrition in critically ill patients with sepsis and acute kidney injury. Curr Opin Nephrol Hypertens 2022; 31:358-366. [PMID: 35703214 PMCID: PMC9248034 DOI: 10.1097/mnh.0000000000000801] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To discuss how nutritional management could be optimized to promote protective metabolism in sepsis and associated acute kidney injury. RECENT FINDINGS Recent evidence suggests that sepsis is a metabolically distinct critical illness and that certain metabolic alterations, such as activation of fasting metabolism, may be protective in bacterial sepsis. These findings may explain the lack of survival benefit in recent randomized controlled trials of nutrition therapy for critical illness. These trials are limited by cohort heterogeneity, combining both septic and nonseptic critical illness, and the use of inaccurate caloric estimates to determine energy requirements. These energy estimates are also unable to provide information on specific substrate preferences or the capacity for substrate utilization. As a result, high protein feeding beyond the capacity for protein synthesis could cause harm in septic patients. Excess glucose and insulin exposures suppress fatty acid oxidation, ketogenesis and autophagy, of which emerging evidence suggest are protective against sepsis associated organ damage such as acute kidney injury. SUMMARY Distinguishing pathogenic and protective sepsis-related metabolic changes are critical to enhancing and individualizing nutrition management for critically ill patients.
Collapse
Affiliation(s)
| | | | - Sarah C Huen
- Department of Internal Medicine
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
24
|
Elajaili H, Hernandez-Lagunas L, Harris P, Sparagna GC, Jonscher R, Ohlstrom D, Sucharov CC, Bowler RP, Suliman H, Fritz KS, Roede JR, Nozik ES. Extracellular superoxide dismutase (EC-SOD) R213G variant reduces mitochondrial ROS and preserves mitochondrial function in bleomycin-induced lung injury: EC-SOD R213G variant and intracellular redox regulation. ADVANCES IN REDOX RESEARCH 2022; 5:100035. [PMID: 38273965 PMCID: PMC10810244 DOI: 10.1016/j.arres.2022.100035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Extracellular superoxide dismutase (EC-SOD) is highly expressed in the lung and vasculature. A common human single nucleotide polymorphism (SNP) in the matrix binding region of EC-SOD leads to a single amino acid substitution, R213G, and alters EC-SOD tissue binding affinity. The change in tissue binding affinity redistributes EC-SOD from tissue to extracellular fluids. Mice (R213G mice) expressing a knock-in of this EC-SOD SNP exhibit elevated plasma and reduced lung EC-SOD content and activity and are protected against bleomycin-induced lung injury and inflammation. It is unknown how the redistribution of EC-SOD alters site-specific redox-regulated molecules relevant for protection. In this study, we tested the hypothesis that the change in the local EC-SOD content would influence not only the extracellular redox microenvironment where EC-SOD is localized but also protect the intracellular redox status of the lung. Mice were treated with bleomycin and harvested 7 days post-treatment. Superoxide levels, measured by electron paramagnetic resonance (EPR), were lower in plasma and Bronchoalveolar lavage fluid (BALF) cells in R213G mice compared to wild-type (WT) mice, while lung cellular superoxide levels in R213G mice were not elevated post-bleomycin compared to WT mice despite low lung EC-SOD levels. Lung glutathione redox potential (EhGSSG), determined by HPLC and fluorescence, was more oxidized in WT compared to R213G mice. In R213G mice, lung mitochondrial oxidative stress was reduced shown by mitochondrial superoxide level measured by EPR in lung and the resistance to bleomycin-induced cardiolipin oxidation. Bleomycin treatment suppressed mitochondrial respiration in WT mice. Mitochondrial function was impaired at baseline in R213G mice but did not exhibit further suppression in respiration post-bleomycin. Collectively, the results indicate that R213G variant preserves intracellular redox state and protects mitochondrial function in the setting of bleomycin-induced inflammation.
Collapse
Affiliation(s)
- Hanan Elajaili
- Cardiovascular Pulmonary Research Laboratories and Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Laura Hernandez-Lagunas
- Cardiovascular Pulmonary Research Laboratories and Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Peter Harris
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Genevieve C. Sparagna
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Raleigh Jonscher
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Denis Ohlstrom
- Cardiovascular Pulmonary Research Laboratories and Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Carmen C. Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Hagir Suliman
- Departments of Anesthesiology and Pathology, Duke University School of Medicine, Durham, North Carolina
| | - Kristofer S. Fritz
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - James R. Roede
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Eva S. Nozik
- Cardiovascular Pulmonary Research Laboratories and Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
25
|
Zou F, Zhuang ZB, Zou SS, Wang B, Zhang ZH. BML-111 alleviates inflammatory response of alveolar epithelial cells via miR-494/Slit2/Robo4 signalling axis to improve acute lung injury. Autoimmunity 2022; 55:318-327. [PMID: 35656971 DOI: 10.1080/08916934.2022.2065671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Fang Zou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, P.R. China
| | - Zhong-Bao Zhuang
- Department of Pharmacy, Hebei North University, Zhangjiakou, Hebei Province, P.R. China
| | - Shuang-Shuang Zou
- Guangzhou Liwan Stomatological Hospital, Guangzhou, Guangdong Province, P.R. China
| | - Bu Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, P.R. China
| | - Zhi-Hua Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei Province, P.R. China
| |
Collapse
|
26
|
Doolittle LM, Binzel K, Nolan KE, Craig K, Rosas LE, Bernier MC, Joseph LM, Woods PS, Knopp MV, Davis IC. CDP-choline Corrects Alveolar Type II Cell Mitochondrial Dysfunction in Influenza-infected Mice. Am J Respir Cell Mol Biol 2022; 66:682-693. [PMID: 35442170 PMCID: PMC9163648 DOI: 10.1165/rcmb.2021-0512oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Development of ARDS in influenza A virus (IAV)-infected mice is associated with inhibition of alveolar type II (ATII) epithelial cell de novo phosphatidylcholine synthesis and administration of the phosphatidylcholine precursor CDP-choline attenuates IAV-induced ARDS in mice. We hypothesized inhibition of phosphatidylcholine synthesis would also impact the function of ATII cell mitochondria. To test this hypothesis, adult C57BL/6 mice of both sexes were inoculated intranasally with 10,000 p.f.u./mouse influenza A/WSN/33 (H1N1). Controls were mock-infected with virus diluent. Mice were treated with saline vehicle or CDP-choline (100 μg/mouse, i.p.) once daily from 1-5 days post-inoculation (dpi). ATII cells were isolated by a standard lung digestion protocol at 6 dpi for analysis of mitochondrial function. IAV infection increased uptake of the glucose analog 18F-FDG by the lungs and caused a switch from oxidative phosphorylation to aerobic glycolysis as a primary means of ATII cell ATP synthesis by 6 dpi. Infection also induced ATII cell mitochondrial depolarization and shrinkage, upregulation of PGC-1α, decreased cardiolipin content, and reduced expression of mitofusin 1, OPA1, DRP1, Complexes I and IV of the electron transport chain, and enzymes involved in cardiolipin synthesis. Daily CDP-choline treatment prevented the declines in oxidative phosphorylation, mitochondrial membrane potential, and cardiolipin synthesis resulting from IAV infection but did not fully reverse the glycolytic shift. CDP-choline also did not prevent the alterations in mitochondrial protein expression resulting from infection. Taken together, our data show ATII cell mitochondrial dysfunction following IAV infection results from impaired de novo phospholipid synthesis, but the glycolytic shift does not.
Collapse
Affiliation(s)
- Lauren M Doolittle
- OHIO STATE UNIVERSITY, COLLEGE OF VETERINARY MEDICINE, Columbus, Ohio, United States
| | - Katherine Binzel
- OHIO STATE UNIVERSITY, Wright Center of Innovation in Biomedical Imaging, Columbus, Ohio, United States
| | - Katherine E Nolan
- The Ohio State University, 2647, Veterinary Biosciences, Columbus, Ohio, United States
| | - Kelsey Craig
- The Ohio State University, 2647, Veterinary Biosciences, Columbus, Ohio, United States
| | - Lucia E Rosas
- The Ohio State University, 2647, Veterinary Biosciences, Columbus, Ohio, United States
| | - Matthew C Bernier
- The Ohio State University, 2647, CCIC Mass Spectrometry & Proteomics Facility, Columbus, Ohio, United States
| | - Lisa M Joseph
- The Ohio State University, 2647, Veterinary Biosciences, Columbus, Ohio, United States
| | - Parker S Woods
- The Ohio State University, 2647, Veterinary Biosciences, Columbus, Ohio, United States
| | - Michael V Knopp
- OHIO STATE UNIVERSITY, Wright Center of Innovation in Biomedical Imaging, Columbus, Ohio, United States
| | - Ian C Davis
- OHIO STATE UNIVERSITY, COLLEGE OF VETERINARY MEDICINE, Columbus, Ohio, United States;
| |
Collapse
|
27
|
Zhang J, Shi J, Han S, Zheng P, Chen Z, Jia G. Titanium dioxide nanoparticles induced reactive oxygen species (ROS) related changes of metabolomics signatures in human normal bronchial epithelial (BEAS-2B) cells. Toxicol Appl Pharmacol 2022; 444:116020. [DOI: 10.1016/j.taap.2022.116020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/13/2022]
|
28
|
Liu J, Yao S, Jia J, Chen Z, Yuan Y, He Y, Wasti B, Duan W, Li D, Wang G, Jia A, Sun W, Qiu S, Ma L, Li J, Liu Y, Zheng J, Xiang X, Zhang X, Liu S, He Z, Peng Z, Zhang H, Zhang D, Xiao B. Loss of MBD2 ameliorates LPS‐induced alveolar epithelial cell apoptosis and ALI in mice via modulating intracellular zinc homeostasis. FASEB J 2022; 36:e22162. [PMID: 35061304 DOI: 10.1096/fj.202100924rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 12/16/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023]
Affiliation(s)
- Jiqiang Liu
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Shuo Yao
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Jingsi Jia
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Zhifeng Chen
- Department of Respiratory Medicine Hunan Center for Evidence‐Based Medicine Research Unit of Respiratory Diseases The Second Xiangya Hospital of Central South University Changsha P.R. China
| | - Yu Yuan
- Department of Respiratory Medicine Hunan Center for Evidence‐Based Medicine Research Unit of Respiratory Diseases The Second Xiangya Hospital of Central South University Changsha P.R. China
| | - Yi He
- Department of Respiratory Medicine Hunan Center for Evidence‐Based Medicine Research Unit of Respiratory Diseases The Second Xiangya Hospital of Central South University Changsha P.R. China
| | - Binaya Wasti
- Department of Respiratory Medicine Hunan Center for Evidence‐Based Medicine Research Unit of Respiratory Diseases The Second Xiangya Hospital of Central South University Changsha P.R. China
| | - Wentao Duan
- Department of Respiratory Medicine Hunan Center for Evidence‐Based Medicine Research Unit of Respiratory Diseases The Second Xiangya Hospital of Central South University Changsha P.R. China
| | - Danhong Li
- Department of Respiratory Medicine Hunan Center for Evidence‐Based Medicine Research Unit of Respiratory Diseases The Second Xiangya Hospital of Central South University Changsha P.R. China
| | - Guyi Wang
- Department of Intensive Care Medicine The Second Xiangya Hospital of Central South University Changsha P.R. China
| | - Aijun Jia
- Department of the Third Emergency of Yuelushan Hospital District Hunan Provincial People's Hospital Changsha P.R. China
| | - Wenjin Sun
- Department of General Medicine West China Hospital, Sichuan University Chengdu P.R. China
| | - Shuangfa Qiu
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Libing Ma
- Department of Respiratory and Critical Care Medicine The Affiliated Hospital of Guilin Medical University Guangxi P.R. China
| | - Jianmin Li
- Department of Respiratory and Critical Care Medicine Hunan Provincial People's Hospital Changsha P.R. China
| | - Yi Liu
- Department of Respiratory Medicine Zhuzhou City Central Hospital Zhuzhou P.R. China
| | - Jianfei Zheng
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Xudong Xiang
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Xiufeng Zhang
- Department of Respiratory Medicine The Second Affiliated Hospital of Hainan Medical University Haikou P.R. China
| | - Shaokun Liu
- Department of Respiratory Medicine Hunan Center for Evidence‐Based Medicine Research Unit of Respiratory Diseases The Second Xiangya Hospital of Central South University Changsha P.R. China
| | - Zhibiao He
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Zhenyu Peng
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Hongliang Zhang
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Dongshan Zhang
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| | - Bing Xiao
- Department of Emergency Medicine The Second Xiangya Hospital of Central South University, Emergency and Difficult Diseases Institute of Central South University Changsha P.R. China
| |
Collapse
|
29
|
Wang M, Wang K, Liao X, Hu H, Chen L, Meng L, Gao W, Li Q. Carnitine Palmitoyltransferase System: A New Target for Anti-Inflammatory and Anticancer Therapy? Front Pharmacol 2021; 12:760581. [PMID: 34764874 PMCID: PMC8576433 DOI: 10.3389/fphar.2021.760581] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 09/17/2021] [Indexed: 11/20/2022] Open
Abstract
Lipid metabolism involves multiple biological processes. As one of the most important lipid metabolic pathways, fatty acid oxidation (FAO) and its key rate-limiting enzyme, the carnitine palmitoyltransferase (CPT) system, regulate host immune responses and thus are of great clinical significance. The effect of the CPT system on different tissues or organs is complex: the deficiency or over-activation of CPT disrupts the immune homeostasis by causing energy metabolism disorder and inflammatory oxidative damage and therefore contributes to the development of various acute and chronic inflammatory disorders and cancer. Accordingly, agonists or antagonists targeting the CPT system may become novel approaches for the treatment of diseases. In this review, we first briefly describe the structure, distribution, and physiological action of the CPT system. We then summarize the pathophysiological role of the CPT system in chronic obstructive pulmonary disease, bronchial asthma, acute lung injury, chronic granulomatous disease, nonalcoholic fatty liver disease, hepatic ischemia–reperfusion injury, kidney fibrosis, acute kidney injury, cardiovascular disorders, and cancer. We are also concerned with the current knowledge in either preclinical or clinical studies of various CPT activators/inhibitors for the management of diseases. These compounds range from traditional Chinese medicines to novel nanodevices. Although great efforts have been made in studying the different kinds of CPT agonists/antagonists, only a few pharmaceuticals have been applied for clinical uses. Nevertheless, research on CPT activation or inhibition highlights the pharmacological modulation of CPT-dependent FAO, especially on different CPT isoforms, as a promising anti-inflammatory/antitumor therapeutic strategy for numerous disorders.
Collapse
Affiliation(s)
- Muyun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kun Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ximing Liao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiyang Hu
- Department of Vascular Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Liangzhi Chen
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linlin Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Gao
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
van Liempd S, Cabrera D, Pilzner C, Kollmus H, Schughart K, Falcón-Pérez JM. Impaired beta-oxidation increases vulnerability to influenza A infection. J Biol Chem 2021; 297:101298. [PMID: 34637789 PMCID: PMC8564733 DOI: 10.1016/j.jbc.2021.101298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/20/2022] Open
Abstract
Influenza A virus (IAV) infection casts a significant burden on society. It has particularly high morbidity and mortality rates in patients suffering from metabolic disorders. The aim of this study was to relate metabolic changes with IAV susceptibility using well-characterized inbred mouse models. We compared the highly susceptible DBA/2J (D2) mouse strain for which IAV infection is lethal with the C57BL/6J (B6) strain, which exhibits a moderate course of disease and survives IAV infection. Previous studies showed that D2 has higher insulin and glucose levels and is predisposed to develop diet-induced type 2 diabetes. Using high-resolution liquid chromatography–coupled MS, the plasma metabolomes of individual animals were repeatedly measured up to 30 days postinfection. The biggest metabolic difference between these strains in healthy and infected states was in the levels of malonylcarnitine, which was consistently increased 5-fold in D2. Other interstrain and intrastrain differences in healthy and infected animals were observed for acylcarnitines, glucose, branched-chain amino acids, and oxidized fatty acids. By mapping metabolic changes to canonical pathways, we found that mitochondrial beta-oxidation is likely disturbed in D2 animals. In noninfected D2 mice, this leads to increased glycerolipid production and reduced acylcarnitine production, whereas in infected D2 animals, peroxisomal beta-oxidation becomes strongly increased. From these studies, we conclude that metabolic changes caused by a distortion of mitochondrial and peroxisomal metabolism might impact the innate immune response in D2, leading to high viral titers and mortality.
Collapse
Affiliation(s)
| | - Diana Cabrera
- Metabolomics Platform CIC bioGUNE-BRTA, Derio, Spain
| | - Carolin Pilzner
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Heike Kollmus
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany; University of Veterinary Medicine Hannover, Hannover, Germany; Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Juan M Falcón-Pérez
- Metabolomics Platform CIC bioGUNE-BRTA, Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
31
|
Hassan AI, Samir A, Youssef HF, Mohamed SS, Asker MS, Mahmoud MG. Effects of silver nanoparticles-polysaccharide on bleomycin-induced pulmonary fibrosis in rats. J Pharm Pharmacol 2021; 73:1503-1512. [PMID: 34515769 DOI: 10.1093/jpp/rgab037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/12/2021] [Indexed: 11/13/2022]
Abstract
OBJECTIVES The first goal of this study was to synthesize the silver nanoparticles Alcaligenes xylosoxidans exopolysaccharide (Ag-AXEPS). The second objective was to analyse the role of Ag-AXEPS nanoparticles (NPS) in treating bleomycin (BLM)-induced lung fibrosis. METHODS Intratracheal bleomycin (2.5 U/kg) was administered to prompt pulmonary fibrosis in rats, and pulmonary fibrosis was treated with Ag-AXEPS nanoparticles (100 ppm/twice a week for four weeks). KEY FINDINGS Ag-AXEPS nanoparticles significantly decreased the diversity of pulmonary inflammatory agents in rats with BLM-induced fibrosis. Reduced levels of respiratory tumor necrosis factor-alpha, monocyte chemotactic protein-1, matrix metalloproteinases (MMP-2 and MMP-9) were observed on treatment with synthesized Ag-AXEPS. Similarly, the treatment decreased IL-12, mRNA levels of BAX and plasma fibrosis markers like N-terminal procollagen III propeptide and transforming growth factor-β1. On the other hand, the treatment increased mRNA BCL2 and total antioxidant capacity. It also lowered the level of fibrosis, as was shown by a quantified pathologic study of hematoxylin-eosin-stained lung parts. The treatment, however, ensured that lung collagen was restored, as assessed by Masson's trichrome stain, and that overall survival was increased and enhanced. CONCLUSIONS Our work showed that nanoparticles could be obtained at 37°C and may be a possible pulmonary fibrosis therapeutic agent.
Collapse
Affiliation(s)
- Amal I Hassan
- Department of Radioisotopes, Nuclear Research Centre, Atomic Energy Authority, Egypt
| | - Amer Samir
- Department of Pathology, National Cancer Institute, Cairo University, Egypt
| | - Hanan F Youssef
- Department of Ceramics, Refractories and Building Materials, National Research Centre, Dokki, Cairo, Egypt
| | - Sahar S Mohamed
- Department of Microbial Biotechnology, National Research Centre, Dokki, Cairo, Egypt
| | - Mohsen S Asker
- Department of Microbial Biotechnology, National Research Centre, Dokki, Cairo, Egypt
| | - Manal G Mahmoud
- Department of Microbial Biotechnology, National Research Centre, Dokki, Cairo, Egypt
| |
Collapse
|
32
|
Prasad K, AlOmar SY, Almuqri EA, Rudayni HA, Kumar V. Genomics-guided identification of potential modulators of SARS-CoV-2 entry proteases, TMPRSS2 and Cathepsins B/L. PLoS One 2021; 16:e0256141. [PMID: 34407143 PMCID: PMC8372896 DOI: 10.1371/journal.pone.0256141] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/31/2021] [Indexed: 01/04/2023] Open
Abstract
SARS-CoV-2 requires serine protease, transmembrane serine protease 2 (TMPRSS2), and cysteine proteases, cathepsins B, L (CTSB/L) for entry into host cells. These host proteases activate the spike protein and enable SARS-CoV-2 entry. We herein performed genomic-guided gene set enrichment analysis (GSEA) to identify upstream regulatory elements altering the expression of TMPRSS2 and CTSB/L. Further, medicinal compounds were identified based on their effects on gene expression signatures of the modulators of TMPRSS2 and CTSB/L genes. Using this strategy, estradiol and retinoic acid have been identified as putative SARS-CoV-2 alleviation agents. Next, we analyzed drug-gene and gene-gene interaction networks using 809 human targets of SARS-CoV-2 proteins. The network results indicate that estradiol interacts with 370 (45%) and retinoic acid interacts with 251 (31%) human proteins. Interestingly, a combination of estradiol and retinoic acid interacts with 461 (56%) of human proteins, indicating the therapeutic benefits of drug combination therapy. Finally, molecular docking analysis suggests that both the drugs bind to TMPRSS2 and CTSL with the nanomolar to low micromolar affinity. The results suggest that these drugs can simultaneously target both the entry pathways of SARS-CoV-2 and thus can be considered as a potential treatment option for COVID-19.
Collapse
Affiliation(s)
- Kartikay Prasad
- Amity Institute of Neuropsychology & Neurosciences (AINN), Amity University, Noida, UP, India
| | - Suliman Yousef AlOmar
- Department of College of Science, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Eman Abdullah Almuqri
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Hassan Ahmed Rudayni
- Biology Department, College of Science, Imam Muhammad bin Saud Islamic University, Riyadh, Kingdom of Saudi Arabia
| | - Vijay Kumar
- Amity Institute of Neuropsychology & Neurosciences (AINN), Amity University, Noida, UP, India
- * E-mail:
| |
Collapse
|
33
|
Chang M, Xu G, Xiong C, Yang X, Yan S, Tao Y, Li H, Li Y, Yao S, Zhao Y. Alpha-lipoic acid attenuates silica-induced pulmonary fibrosis by improving mitochondrial function via AMPK/PGC1α pathway activation in C57BL/6J mice. Toxicol Lett 2021; 350:121-132. [PMID: 34252510 DOI: 10.1016/j.toxlet.2021.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 05/22/2021] [Accepted: 07/07/2021] [Indexed: 11/17/2022]
Abstract
Silicosis is characterized by pulmonary interstitial fibrosis that arises as a result of chronic exposure to silica. The few available treatments only delay its progression. As α-lipoic acid (ALA) has been shown to have various beneficial effects, including mitoprotective, antioxidant, and anti-inflammatory effects, we hypothesized that it may exhibit therapeutic effects in pulmonary fibrosis. Therefore, in the present study, we used a murine model of silicosis to investigate whether supplementation with exogenous ALA could attenuate silica-induced pulmonary fibrosis by improving mitochondrial function. ALA was administered to the model mice via continuous intragastric administration for 28 days, and then the antioxidant and mitoprotective effects of ALA were evaluated. The results showed that ALA decreased the production of reactive oxygen species, protected mitochondria from silica-induced dysfunction, and inhibited extracellular matrix deposition. ALA also decreased hyperglycemia and hyperlipidemia. Activation of the mitochondrial AMPK/PGC1α pathway might be responsible for these ALA-mediated anti-fibrotic effects. Exogenous ALA blocked oxidative stress by activating NRF2. Taken together, these findings demonstrate that exogenous ALA effectively prevents the progression of silicosis in a murine model, likely by stimulating mitochondrial biogenesis and endogenous antioxidant responses. Therefore, ALA can potentially delay the progression of silica-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Meiyu Chang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Guangcui Xu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Cheng Xiong
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Xuesi Yang
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Sensen Yan
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Yingjun Tao
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Haibin Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Yuchun Li
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Sanqiao Yao
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China
| | - Yingzheng Zhao
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan Province, 453003, PR China.
| |
Collapse
|
34
|
Cinar R, Park JK, Zawatsky CN, Coffey NJ, Bodine SP, Abdalla J, Yokoyama T, Jourdan T, Jay L, Zuo MXG, O'Brien KJ, Huang J, Mackie K, Alimardanov A, Iyer MR, Gahl WA, Kunos G, Gochuico BR, Malicdan MCV. CB 1 R and iNOS are distinct players promoting pulmonary fibrosis in Hermansky-Pudlak syndrome. Clin Transl Med 2021; 11:e471. [PMID: 34323400 PMCID: PMC8255071 DOI: 10.1002/ctm2.471] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
Hermansky-Pudlak syndrome (HPS) is a rare genetic disorder which, in its most common and severe form, HPS-1, leads to fatal adult-onset pulmonary fibrosis (PF) with no effective treatment. We evaluated the role of the endocannabinoid/CB1 R system and inducible nitric oxide synthase (iNOS) for dual-target therapeutic strategy using human bronchoalveolar lavage fluid (BALF), lung samples from patients with HPS and controls, HPS-PF patient-derived lung fibroblasts, and bleomycin-induced PF in pale ear mice (HPS1ep/ep ). We found overexpression of CB1 R and iNOS in fibrotic lungs of HPSPF patients and bleomycin-infused pale ear mice. The endocannabinoid anandamide was elevated in BALF and negatively correlated with pulmonary function parameters in HPSPF patients and pale ear mice with bleomycin-induced PF. Simultaneous targeting of CB1 R and iNOS by MRI-1867 yielded greater antifibrotic efficacy than inhibiting either target alone by attenuating critical pathologic pathways. Moreover, MRI-1867 treatment abrogated bleomycin-induced increases in lung levels of the profibrotic interleukin-11 via iNOS inhibition and reversed mitochondrial dysfunction via CB1 R inhibition. Dual inhibition of CB1 R and iNOS is an effective antifibrotic strategy for HPSPF.
Collapse
Affiliation(s)
- Resat Cinar
- Section on Fibrotic DisordersNational Institute on Alcohol Abuse and Alcoholism, National Institutes of HealthMarylandUSA
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Joshua K. Park
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Charles N. Zawatsky
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Nathan J. Coffey
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Steven P. Bodine
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Jasmina Abdalla
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Tadafumi Yokoyama
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
- Present address:
Department of PediatricsKanazawa UniversityKanazawaJapan
| | - Tony Jourdan
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
- Present address:
INSERM Lipids, Nutrition, Cancer UMR1231University of Burgundy and Franche‐ComtéDijonFrance
| | - Lindsey Jay
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Mei Xing G. Zuo
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Kevin J. O'Brien
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Junfeng Huang
- Therapeutics Development BranchDivision of Preclinical InnovationNational Center for Advancing Translational SciencesNational Institutes of HealthRockvilleMarylandUSA
| | - Ken Mackie
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonIndianaUSA
| | - Asaf Alimardanov
- Therapeutics Development BranchDivision of Preclinical InnovationNational Center for Advancing Translational SciencesNational Institutes of HealthRockvilleMarylandUSA
| | - Malliga R. Iyer
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - William A. Gahl
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
- NIH Undiagnosed Diseases Program and Office of the Clinical DirectorNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - George Kunos
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMarylandUSA
| | - Bernadette R. Gochuico
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - May Christine V. Malicdan
- Section of Human Biochemical GeneticsMedical Genetics BranchNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
- NIH Undiagnosed Diseases Program and Office of the Clinical DirectorNational Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
35
|
Genetic drivers of m 6A methylation in human brain, lung, heart and muscle. Nat Genet 2021; 53:1156-1165. [PMID: 34211177 DOI: 10.1038/s41588-021-00890-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 05/18/2021] [Indexed: 01/22/2023]
Abstract
The most prevalent post-transcriptional mRNA modification, N6-methyladenosine (m6A), plays diverse RNA-regulatory roles, but its genetic control in human tissues remains uncharted. Here we report 129 transcriptome-wide m6A profiles, covering 91 individuals and 4 tissues (brain, lung, muscle and heart) from GTEx/eGTEx. We integrate these with interindividual genetic and expression variation, revealing 8,843 tissue-specific and 469 tissue-shared m6A quantitative trait loci (QTLs), which are modestly enriched in, but mostly orthogonal to, expression QTLs. We integrate m6A QTLs with disease genetics, identifying 184 GWAS-colocalized m6A QTL, including brain m6A QTLs underlying neuroticism, depression, schizophrenia and anxiety; lung m6A QTLs underlying expiratory flow and asthma; and muscle/heart m6A QTLs underlying coronary artery disease. Last, we predict novel m6A regulators that show preferential binding in m6A QTLs, protein interactions with known m6A regulators and expression correlation with the m6A levels of their targets. Our results provide important insights and resources for understanding both cis and trans regulation of epitranscriptomic modifications, their interindividual variation and their roles in human disease.
Collapse
|
36
|
AbdelMassih AF, Menshawey R, Ismail JH, Husseiny RJ, Husseiny YM, Yacoub S, Kamel A, Hozaien R, Yacoub E, Menshawey E, Abdelmalek A, Abouelazaem A, Elhatw A, Aboelmaaty A, Shahib A, Mansour A, Kamal A, Mohamed B, Atif B, Ghabreal B, Abdelmalak C, Ibrahim D, Elsaify E, Magdy F, Hanna FG, Hafez H, Dahir H, Merhom K, Ahmed M, Bishara M, Tawfik M, Youssef M, El Sharnouby M, Hamouda M, Ammar M, Ali N, Daniel N, El-Husseiny N, Abdelraouf N, Abdelhameed NK, Ahmed R, Othman R, Mohamadein R, Allam R, Elgendy R, Shebl R, Elsherbiney S, Fouad S, Emel S, Owais S, Hetta S, El-Saman S, Abdelalim S, Galal S, Asar Y, Osman Y, Khalaf Y, Aziz Y, Khafagy Y, Gamal N, Castaldi B. PPAR agonists as effective adjuvants for COVID-19 vaccines, by modifying immunogenetics: a review of literature. J Genet Eng Biotechnol 2021; 19:82. [PMID: 34057580 PMCID: PMC8165506 DOI: 10.1186/s43141-021-00179-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/14/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Several coronavirus vaccine have been fast-tracked to halt the pandemic, the usage of immune adjuvants that can boost immunological memory has come up to the surface. This is particularly of importance in view of the rates of failure of seroconversion and re-infection after COVID-19 infection, which could make the vaccine role and response debatable. Peroxisome proliferator-activated receptors (PPARs) have an established immune-modulatory role, but their effects as adjuvants to vaccination have not been explored to date. It is increasingly recognized that PPAR agonists can upregulate the levels of anti-apoptotic factors such as MCL-1. Such effect can improve the results of vaccination by enhancing the longevity of long-lived plasma cells (LLPCs). The interaction between PPAR agonists and the immune system does not halt here, as T cell memory is also stimulated through enhanced T regulatory cells, antagonizing PD-L1 and switching the metabolism of T cells to fatty acid oxidation, which has a remarkable effect on the persistence of T memory cells. What is even of a more significant value is the effect of PPAR gamma on ensuring a profound secretion of antibodies upon re-exposure to the offending antigen through upregulating lipoxin B4, therefore potentially assisting the vaccine response and deterring re-infection. SHORT CONCLUSION In view of the above, we suggest the use of PPAR as adjuvants to vaccines in general especially the emerging COVID-19 vaccine due to their role in enhancing immunologic memory through DNA-dependent mechanisms.
Collapse
Affiliation(s)
- Antoine Fakhry AbdelMassih
- Pediatric Cardiology Unit, Pediatrics' Department, Cairo University Children Hospital, Faculty of Medicine, Cairo University, Kasr Al Ainy Street, Cairo, 12411, Egypt.
- Pediatric Cardio-Oncology Department, Children Cancer Hospital of Egypt (57357), Cairo, Egypt.
| | - Rahma Menshawey
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Jumana H Ismail
- Pulmonology Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Reem J Husseiny
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Yousef M Husseiny
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, New Giza University, 6th of October City, Egypt
| | - Shenoda Yacoub
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Aya Kamel
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Rafeef Hozaien
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Elaria Yacoub
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Esraa Menshawey
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Abanoub Abdelmalek
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Ahmed Abouelazaem
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Ahmed Elhatw
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Ahmed Aboelmaaty
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Alaaelrahman Shahib
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Amany Mansour
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Aya Kamal
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Basant Mohamed
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Bemen Atif
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Beshoy Ghabreal
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Catherine Abdelmalak
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - David Ibrahim
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Ebtesam Elsaify
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Farah Magdy
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Farid G Hanna
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Hadeer Hafez
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Hafsa Dahir
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Kerlos Merhom
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Maram Ahmed
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Mariam Bishara
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Mina Tawfik
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Mina Youssef
- University at Buffalo School of Medicine and Biomedical, Buffalo, USA
| | - Mohamed El Sharnouby
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Mourad Hamouda
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Musheera Ammar
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Nada Ali
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Nada Daniel
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nadine El-Husseiny
- Faculty of Dentistry, Cairo University, Giza, Egypt
- Pixagon graphic design Agency, Cairo, Egypt
| | - Noha Abdelraouf
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Nuran K Abdelhameed
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Radwa Ahmed
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Radwa Othman
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Rahma Mohamadein
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Rana Allam
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Rana Elgendy
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Rana Shebl
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Saged Elsherbiney
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Sarah Fouad
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Sara Emel
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Sara Owais
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Sarah Hetta
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Samah El-Saman
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Shaimaa Abdelalim
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherin Galal
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Yara Asar
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Yara Osman
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Yasmeen Khalaf
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Youstina Aziz
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, 6th October University, 6th of October City, Egypt
| | - Yousra Khafagy
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Nervana Gamal
- Research Accessibility Team, Student and Internship research program, Faculty of Medicine, Cairo University, Giza, Egypt
| | | |
Collapse
|
37
|
Shi D, Yan R, Lv L, Jiang H, Lu Y, Sheng J, Xie J, Wu W, Xia J, Xu K, Gu S, Chen Y, Huang C, Guo J, Du Y, Li L. The serum metabolome of COVID-19 patients is distinctive and predictive. Metabolism 2021; 118:154739. [PMID: 33662365 PMCID: PMC7920809 DOI: 10.1016/j.metabol.2021.154739] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Metabolism is critical for sustaining life, immunity and infection, but its role in COVID-19 is not fully understood. METHODS Seventy-nine COVID-19 patients, 78 healthy controls (HCs) and 30 COVID-19-like patients were recruited in a prospective cohort study. Samples were collected from COVID-19 patients with mild or severe symptoms on admission, patients who progressed from mild to severe symptoms, and patients who were followed from hospital admission to discharge. The metabolome was assayed using gas chromatography-mass spectrometry. RESULTS Serum butyric acid, 2-hydroxybutyric acid, l-glutamic acid, l-phenylalanine, l-serine, l-lactic acid, and cholesterol were enriched in COVID-19 and COVID-19-like patients versus HCs. Notably, d-fructose and succinic acid were enriched, and citric acid and 2-palmitoyl-glycerol were depleted in COVID-19 patients compared to COVID-19-like patients and HCs, and these four metabolites were not differentially distributed in non-COVID-19 groups. COVID-19 patients had enriched 4-deoxythreonic acid and depleted 1,5-anhydroglucitol compared to HCs and enriched oxalic acid and depleted phosphoric acid compared to COVID-19-like patients. A combination of d-fructose, citric acid and 2-palmitoyl-glycerol distinguished COVID-19 patients from HCs and COVID-19-like patients, with an area under the curve (AUC) > 0.92 after validation. The combination of 2-hydroxy-3-methylbutyric acid, 3-hydroxybutyric acid, cholesterol, succinic acid, L-ornithine, oleic acid and palmitelaidic acid predicted patients who progressed from mild to severe COVID-19, with an AUC of 0.969. After discharge, nearly one-third of metabolites were recovered in COVID-19 patients. CONCLUSIONS The serum metabolome of COVID-19 patients is distinctive and has important value in investigating pathogenesis, determining a diagnosis, predicting severe cases, and improving treatment.
Collapse
Affiliation(s)
- Ding Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Ren Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Huiyong Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yingfeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jifang Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jiaojiao Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Wenrui Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jiafeng Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Kaijin Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Silan Gu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Chenjie Huang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jing Guo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yiling Du
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China; Institute of Pharmaceutical Biotechnology and The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
38
|
Varon J, Englert JA. Kidney-lung cross talk during ARDS: mitochondrial DAMPs join the conversation. Am J Physiol Lung Cell Mol Physiol 2021; 320:L819-L820. [PMID: 33688748 PMCID: PMC8384563 DOI: 10.1152/ajplung.00093.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Jack Varon
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joshua A Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
39
|
Yang CC, Yang CM. Chinese Herbs and Repurposing Old Drugs as Therapeutic Agents in the Regulation of Oxidative Stress and Inflammation in Pulmonary Diseases. J Inflamm Res 2021; 14:657-687. [PMID: 33707963 PMCID: PMC7940992 DOI: 10.2147/jir.s293135] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Several pro-inflammatory factors and proteins have been characterized that are involved in the pathogenesis of inflammatory diseases, including acute respiratory distress syndrome, chronic obstructive pulmonary disease, and asthma, induced by oxidative stress, cytokines, bacterial toxins, and viruses. Reactive oxygen species (ROS) act as secondary messengers and are products of normal cellular metabolism. Under physiological conditions, ROS protect cells against oxidative stress through the maintenance of cellular redox homeostasis, which is important for proliferation, viability, cell activation, and organ function. However, overproduction of ROS is most frequently due to excessive stimulation of either the mitochondrial electron transport chain and xanthine oxidase or reduced nicotinamide adenine dinucleotide phosphate (NADPH) by pro-inflammatory cytokines, such as interleukin-1β and tumor necrosis factor α. NADPH oxidase activation and ROS overproduction could further induce numerous inflammatory target proteins that are potentially mediated via Nox/ROS-related transcription factors triggered by various intracellular signaling pathways. Thus, oxidative stress is considered important in pulmonary inflammatory processes. Previous studies have demonstrated that redox signals can induce pulmonary inflammatory diseases. Thus, therapeutic strategies directly targeting oxidative stress may be effective for pulmonary inflammatory diseases. Therefore, drugs with anti-inflammatory and anti-oxidative properties may be beneficial to these diseases. Recent studies have suggested that traditional Chinese medicines, statins, and peroxisome proliferation-activated receptor agonists could modulate inflammation-related signaling processes and may be beneficial for pulmonary inflammatory diseases. In particular, several herbal medicines have attracted attention for the management of pulmonary inflammatory diseases. Therefore, we reviewed the pharmacological effects of these drugs to dissect how they induce host defense mechanisms against oxidative injury to combat pulmonary inflammation. Moreover, the cytotoxicity of oxidative stress and apoptotic cell death can be protected via the induction of HO-1 by these drugs. The main objective of this review is to focus on Chinese herbs and old drugs to develop anti-inflammatory drugs able to induce HO-1 expression for the management of pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Tao-Yuan, Kwei-San, Tao-Yuan, 33302, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, 33302, Taiwan
| | - Chuen-Mao Yang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, 40402, Taiwan.,Ph.D. Program for Biotech Pharmaceutical Industry, China Medical University, Taichung, 40402, Taiwan.,Department of Post-Baccalaureate Veterinary Medicine, College of Medical and Health Science, Asia University, Taichung, 41354, Taiwan
| |
Collapse
|
40
|
Khan MAAK, Islam ABMMK. SARS-CoV-2 Proteins Exploit Host's Genetic and Epigenetic Mediators for the Annexation of Key Host Signaling Pathways. Front Mol Biosci 2021; 7:598583. [PMID: 33585554 PMCID: PMC7872968 DOI: 10.3389/fmolb.2020.598583] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
The constant rise of the death toll and cases of COVID-19 has made this pandemic a serious threat to human civilization. Understanding of host-SARS-CoV-2 interaction in viral pathogenesis is still in its infancy. In this study, we utilized a blend of computational and knowledgebase approaches to model the putative virus-host interplay in host signaling pathways by integrating the experimentally validated host interactome proteins and differentially expressed host genes in SARS-CoV-2 infection. While searching for the pathways in which viral proteins interact with host proteins, we discovered various antiviral immune response pathways such as hypoxia-inducible factor 1 (HIF-1) signaling, autophagy, retinoic acid-inducible gene I (RIG-I) signaling, Toll-like receptor signaling, fatty acid oxidation/degradation, and IL-17 signaling. All these pathways can be either hijacked or suppressed by the viral proteins, leading to improved viral survival and life cycle. Aberration in pathways such as HIF-1 signaling and relaxin signaling in the lungs suggests the pathogenic lung pathophysiology in COVID-19. From enrichment analysis, it was evident that the deregulated genes in SARS-CoV-2 infection might also be involved in heart development, kidney development, and AGE-RAGE signaling pathway in diabetic complications. Anomalies in these pathways might suggest the increased vulnerability of COVID-19 patients with comorbidities. Moreover, we noticed several presumed infection-induced differentially expressed transcription factors and epigenetic factors, such as miRNAs and several histone modifiers, which can modulate different immune signaling pathways, helping both host and virus. Our modeling suggests that SARS-CoV-2 integrates its proteins in different immune signaling pathways and other cellular signaling pathways for developing efficient immune evasion mechanisms while leading the host to a more complicated disease condition. Our findings would help in designing more targeted therapeutic interventions against SARS-CoV-2.
Collapse
|
41
|
Wang H, Sun X, Lu Q, Zemskov EA, Yegambaram M, Wu X, Wang T, Tang H, Black SM. The mitochondrial redistribution of eNOS is involved in lipopolysaccharide induced inflammasome activation during acute lung injury. Redox Biol 2021; 41:101878. [PMID: 33578126 PMCID: PMC7879038 DOI: 10.1016/j.redox.2021.101878] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/17/2021] [Accepted: 01/21/2021] [Indexed: 01/03/2023] Open
Abstract
Acute lung injury (ALI) is a devastating clinical syndrome with no effective therapies. Inflammasome activation has been reported to play a critical role in the initiation and progression of ALI. The molecular mechanisms involved in regulating the activation of inflammasome in ALI remains unresolved, although increases in mitochondrial derived reactive oxygen species (mito-ROS) are involved. Our previous work has shown that the mitochondrial redistribution of uncoupled eNOS impairs mitochondrial bioenergetics and increases mito-ROS generation. Thus, the focus of our study was to determine if lipopolysaccharide (LPS)-mediated inflammasome activation involves the mitochondrial redistribution of uncoupled eNOS. Our data show that the increase in mito-ROS involved in LPS-mediated inflammasome activation is associated with the disruption of mitochondrial bioenergetics in human lung microvascular endothelial cells (HLMVEC) and the mitochondrial redistribution of eNOS. These effects are dependent on RhoA-ROCK signaling and are mediated via increased phosphorylation of eNOS at Threonine (T)-495. A derivative of the mitochondrial targeted Szeto-Schiller peptide (SSP) attached to the antioxidant Tiron (T-SSP), significantly attenuated LPS-mediated mito-ROS generation and inflammasome activation in HLMVEC. Further, T-SSP attenuated mitochondrial superoxide production in a mouse model of sepsis induced ALI. This in turn significantly reduced the inflammatory response and attenuated lung injury. Thus, our findings show that the mitochondrial redistribution of uncoupled eNOS is intimately involved in the activation of the inflammatory response in ALI and implicate attenuating mito-ROS as a therapeutic strategy in humans.
Collapse
Affiliation(s)
- Hui Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xutong Sun
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Qing Lu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Evgeny A Zemskov
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Manivannan Yegambaram
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xiaomin Wu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, AZ, USA
| | - Haiyang Tang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China; Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA.
| | - Stephen M Black
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
42
|
Hamanaka RB, Mutlu GM. Metabolic requirements of pulmonary fibrosis: role of fibroblast metabolism. FEBS J 2021; 288:6331-6352. [PMID: 33393204 DOI: 10.1111/febs.15693] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/22/2020] [Accepted: 12/31/2020] [Indexed: 12/27/2022]
Abstract
Fibrosis is a pathologic condition characterized by excessive deposition of extracellular matrix and chronic scaring that can affect every organ system. Organ fibrosis is associated with significant morbidity and mortality, contributing to as many as 45% of all deaths in the developed world. In the lung, many chronic lung diseases may lead to fibrosis, the most devastating being idiopathic pulmonary fibrosis (IPF), which affects approximately 3 million people worldwide and has a median survival of 3.8 years. Currently approved therapies for IPF do not significantly extend lifespan, and thus, there is pressing need for novel therapeutic strategies to treat IPF and other fibrotic diseases. At the heart of pulmonary fibrosis are myofibroblasts, contractile cells with characteristics of both fibroblasts and smooth muscle cells, which are the primary cell type responsible for matrix deposition in fibrotic diseases. Much work has centered around targeting the extracellular growth factors and intracellular signaling regulators of myofibroblast differentiation. Recently, metabolic changes associated with myofibroblast differentiation have come to the fore as targetable mechanisms required for myofibroblast function. In this review, we will discuss the metabolic changes associated with myofibroblast differentiation, as well as the mechanisms by which these changes promote myofibroblast function. We will then discuss the potential for this new knowledge to lead to the development of novel therapies for IPF and other fibrotic diseases.
Collapse
Affiliation(s)
- Robert B Hamanaka
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, IL, USA
| | - Gökhan M Mutlu
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, IL, USA
| |
Collapse
|
43
|
Lu Q, Zemskov EA, Sun X, Wang H, Yegambaram M, Wu X, Garcia-Flores A, Song S, Tang H, Kangath A, Cabanillas GZ, Yuan JXJ, Wang T, Fineman JR, Black SM. Activation of the mechanosensitive Ca 2+ channel TRPV4 induces endothelial barrier permeability via the disruption of mitochondrial bioenergetics. Redox Biol 2021; 38:101785. [PMID: 33221570 PMCID: PMC7691184 DOI: 10.1016/j.redox.2020.101785] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/20/2022] Open
Abstract
Mechanical ventilation is a life-saving intervention in critically ill patients with respiratory failure due to acute respiratory distress syndrome (ARDS), a refractory lung disease with an unacceptable high mortality rate. Paradoxically, mechanical ventilation also creates excessive mechanical stress that directly augments lung injury, a syndrome known as ventilator-induced lung injury (VILI). The specific mechanisms involved in VILI-induced pulmonary capillary leakage, a key pathologic feature of VILI are still far from resolved. The mechanoreceptor, transient receptor potential cation channel subfamily V member 4, TRPV4 plays a key role in the development of VILI through unresolved mechanism. Endothelial nitric oxide synthase (eNOS) uncoupling plays an important role in sepsis-mediated ARDS so in this study we investigated whether there is a role for eNOS uncoupling in the barrier disruption associated with TRPV4 activation during VILI. Our data indicate that the TRPV4 agonist, 4α-Phorbol 12,13-didecanoate (4αPDD) induces pulmonary arterial endothelial cell (EC) barrier disruption through the disruption of mitochondrial bioenergetics. Mechanistically, this occurs via the mitochondrial redistribution of uncoupled eNOS secondary to a PKC-dependent phosphorylation of eNOS at Threonine 495 (T495). A specific decoy peptide to prevent T495 phosphorylation reduced eNOS uncoupling and mitochondrial redistribution and preserved PAEC barrier function under 4αPDD challenge. Further, our eNOS decoy peptide was able to preserve lung vascular integrity in a mouse model of VILI. Thus, we have revealed a functional link between TRPV4 activation, PKC-dependent eNOS phosphorylation at T495, and EC barrier permeability. Reducing pT495-eNOS could be a new therapeutic approach for the prevention of VILI.
Collapse
Affiliation(s)
- Qing Lu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Evgeny A Zemskov
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xutong Sun
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Hui Wang
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Manivannan Yegambaram
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Xiaomin Wu
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Alejandro Garcia-Flores
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Shanshan Song
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Haiyang Tang
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA; College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Archana Kangath
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA
| | - Gabriela Zubiate Cabanillas
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA; Department of Chemist-Biological Sciences, Universidad de Sonora, Hermosillo, SON, Mexico
| | - Jason X-J Yuan
- Department of Medicine, University of California, San Diego, CA, USA
| | - Ting Wang
- Department of Internal Medicine, The University of Arizona Health Sciences, Phoenix, AZ, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA; Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Stephen M Black
- Department of Medicine, Division of Translational & Regenerative Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
44
|
Sherren PB, Ostermann M, Agarwal S, Meadows CIS, Ioannou N, Camporota L. COVID-19-related organ dysfunction and management strategies on the intensive care unit: a narrative review. Br J Anaesth 2020; 125:912-925. [PMID: 32988604 PMCID: PMC7833857 DOI: 10.1016/j.bja.2020.08.050] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/17/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has resulted in a significant surge of critically ill patients and an unprecedented demand on intensive care services. The rapidly evolving understanding of pathogenesis, limited disease specific evidence, and demand-resource imbalances have posed significant challenges for intensive care clinicians. COVID-19 is a complex multisystem inflammatory vasculopathy with a significant mortality implication for those admitted to intensive care. Institutional strategic preparation and meticulous intensive care support are essential to maximising outcomes during the pandemic. The significant mortality variation observed between institutions and internationally, despite a single aetiology and uniform presentation, highlights the potential influence of management strategies on outcome. Given that optimal organ support and adjunctive therapies for COVID-19 have not yet been well defined by trial-based outcomes, strategies are predicated on existing literature and experiential learning. This review outlines the relevant pathophysiology and management strategies for critically ill patients with COVID-19, and shares some of the collective learning accumulated in a high volume severe respiratory failure centre in London.
Collapse
Affiliation(s)
| | | | - Sangita Agarwal
- Department of Rheumatology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | | | | |
Collapse
|
45
|
Jiang D, Cui H, Xie N, Banerjee S, Liu RM, Dai H, Thannickal VJ, Liu G. ATF4 Mediates Mitochondrial Unfolded Protein Response in Alveolar Epithelial Cells. Am J Respir Cell Mol Biol 2020; 63:478-489. [PMID: 32551949 DOI: 10.1165/rcmb.2020-0107oc] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Although endoplasmic reticulum (ER) unfolded protein response (UPRER) is well known, mitochondrial unfolded protein response (UPRmt) has not been recognized in alveolar epithelial cells. Furthermore, ER stress and mitochondrial dysfunction are frequently encountered in alveolar epithelial cells from an array of lung disorders. However, these two scenarios have been often regarded as separate mechanisms contributing to the pathogeneses. It is unclear whether there is interplay between these two phenomena or an integrator that couples these two signaling cascades in the stressed alveolar epithelial cells from those pathologies. In this study, we defined UPRmt in alveolar epithelial cells and identified ATF4 (activating transcription factor 4), but not ATF5, as the key regulator of UPRmt. We found that UPRER led to UPRmt and mitochondrial dysfunction in an ATF4-dependent manner. In contrast, mitochondrial stresses did not activate UPRER. We found that alveolar epithelial ATF4 and UPRmt were induced in aged mice with experimental pulmonary fibrosis as well as in patients with idiopathic pulmonary fibrosis. Finally, we found that the inducible expression of ATF4 in mouse alveolar epithelial cells aggravated pulmonary UPRmt, lung inflammation, body weight loss, and death upon bleomycin-induced lung injury. In conclusion, ER stress induces ATF4-dependent UPRmt and mitochondrial dysfunction, indicating a novel mechanism by which ER stress contributes to the pathogeneses of a variety of pulmonary disorders.
Collapse
Affiliation(s)
- Dingyuan Jiang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and.,Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Huachun Cui
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Na Xie
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Sami Banerjee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, National Clinical Research Center for Respiratory Diseases, Beijing, China
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Gang Liu
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
46
|
Zhao X, Gu C, Wang Y. PAD4 selective inhibitor TDFA protects lipopolysaccharide-induced acute lung injury by modulating nuclear p65 localization in epithelial cells. Int Immunopharmacol 2020; 88:106923. [PMID: 32889238 DOI: 10.1016/j.intimp.2020.106923] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/16/2020] [Accepted: 08/18/2020] [Indexed: 11/26/2022]
Abstract
Protein arginine deiminase 4 (PAD4) serves a critical role in differentiation, development and apoptosis through gene regulation and has emerged as a potential therapeutic target for the treatment of various diseases. However, the roles of PAD4 in lipopolysaccharide (LPS)-induced acute lung injury (ALI) remain largely unknown. To investigate the roles of PAD4 during LPS-induced ALI, the present study detected the trend of PAD4 expression in the lung tissues of ALI mice. Subsequently, the efficiency of TDFA on PAD4 and citrullinated H3 histone were detected. And then, histology, the wet/dry weight ratio, survival rate, activated cells infiltration, oxidative stress levels, tight junction proteins and proinflammatory cytokine expression were detected. In addition, the level of transepithelial electrical resistance (TEER) was assessed. Finally, the level of nuclear P65, total phosphorylated P65 and P65 were measured in vivo and in vitro. The results showed that PAD4 expression was upregulated in the lung tissues of LPS-induced ALI. TDFA efficiently decreased the severity of the lung edema, attenuated the severity of pulmonary injury and improved the survival rate following lethal LPS administration. Besides, TDFA reduced activated cells infiltration and suppressed inflammation related parameters, including proinflammatory cytokines production (TNF-α, IL-6 and IL-1β) and oxidative stress (MDA, GSH and SOD). Furthermore, TDFA reversed the TEER downregulation tendency and tight junction proteins (ZO-1, Occludin, Claudin-4) levels that represent the integrity of alveolar epithelium. Eventually, TDFA exerts its protective roles through modulating nuclear localization of transcription factor NF-κB P65 in epithelial cells. Taken together, these results indicate that PAD4 inhibition may serve as a promising therapeutic approach for LPS-induced ALI.
Collapse
Affiliation(s)
- Xiaohong Zhao
- Department of Anesthesia and Perioperative Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250014, PR China; Department of Anesthesiology, The 960(th) Hospital of the People's Liberation Army of China, Jinan, Shandong 250031, PR China
| | - Changping Gu
- Department of Anesthesia and Perioperative Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250014, PR China
| | - Yuelan Wang
- Department of Anesthesia and Perioperative Medicine, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250014, PR China.
| |
Collapse
|
47
|
Heffernan KS, Ranadive SM, Jae SY. Exercise as medicine for COVID-19: On PPAR with emerging pharmacotherapy. Med Hypotheses 2020; 143:110197. [PMID: 33017906 PMCID: PMC7430295 DOI: 10.1016/j.mehy.2020.110197] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022]
Abstract
Coronavirus disease 2019 (COVID-19) may have a metabolic origin given strong links with risk factors such as lipids and glucose and co-morbidities such as obesity and type 2 diabetes mellitus. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein mediates viral cellular entry via the ACE2 receptor. The cytoplasmic tail of this spike protein is heavily palmitoylated. Emerging studies suggest that SARS-CoV-2 alters lipid metabolism in the lung epithelial cells by modulating peroxisome proliferator-activated receptor alpha (PPARα), possibly contributing to lipotoxicity, inflammation and untoward respiratory effects. Disruption of this process may affect palmitoylation of SARS-CoV spike protein and thus infectivity and viral assembly. COVID-19 is also increasingly being recognized as a vascular disease, with several studies noting prominent systemic endothelial dysfunction. The pathogenesis of endothelial dysfunction may also be linked to COVID-19-mediated metabolic and inflammatory effects. Herein, exercise will be compared to fenofibrate as a possible therapeutic strategy to bolster resilience against (and help manage recovery from) COVID-19. This paper will explore the hypothesis that exercise may be a useful adjuvant in a setting of COVID-19 management/rehabilitation due to its effects on PPARα and vascular endothelial function.
Collapse
Affiliation(s)
- Kevin S Heffernan
- Department of Exercise Science, Syracuse University, Syracuse, NY, USA.
| | | | - Sae Young Jae
- Department of Sports Sciences, University of Seoul, Seoul, South Korea
| |
Collapse
|
48
|
COVID-19: Proposing a Ketone-Based Metabolic Therapy as a Treatment to Blunt the Cytokine Storm. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6401341. [PMID: 33014275 PMCID: PMC7519203 DOI: 10.1155/2020/6401341] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Human SARS-CoV-2 infection is characterized by a high mortality rate due to some patients developing a large innate immune response associated with a cytokine storm and acute respiratory distress syndrome (ARDS). This is characterized at the molecular level by decreased energy metabolism, altered redox state, oxidative damage, and cell death. Therapies that increase levels of (R)-beta-hydroxybutyrate (R-BHB), such as the ketogenic diet or consuming exogenous ketones, should restore altered energy metabolism and redox state. R-BHB activates anti-inflammatory GPR109A signaling and inhibits the NLRP3 inflammasome and histone deacetylases, while a ketogenic diet has been shown to protect mice from influenza virus infection through a protective γδ T cell response and by increasing electron transport chain gene expression to restore energy metabolism. During a virus-induced cytokine storm, metabolic flexibility is compromised due to increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) that damage, downregulate, or inactivate many enzymes of central metabolism including the pyruvate dehydrogenase complex (PDC). This leads to an energy and redox crisis that decreases B and T cell proliferation and results in increased cytokine production and cell death. It is hypothesized that a moderately high-fat diet together with exogenous ketone supplementation at the first signs of respiratory distress will increase mitochondrial metabolism by bypassing the block at PDC. R-BHB-mediated restoration of nucleotide coenzyme ratios and redox state should decrease ROS and RNS to blunt the innate immune response and the associated cytokine storm, allowing the proliferation of cells responsible for adaptive immunity. Limitations of the proposed therapy include the following: it is unknown if human immune and lung cell functions are enhanced by ketosis, the risk of ketoacidosis must be assessed prior to initiating treatment, and permissive dietary fat and carbohydrate levels for exogenous ketones to boost immune function are not yet established. The third limitation could be addressed by studies with influenza-infected mice. A clinical study is warranted where COVID-19 patients consume a permissive diet combined with ketone ester to raise blood ketone levels to 1 to 2 mM with measured outcomes of symptom severity, length of infection, and case fatality rate.
Collapse
|
49
|
Arabi YM, Mallampalli R, Englert JA, Bosch NA, Walkey AJ, Al-Dorzi HM. Update in Critical Care 2019. Am J Respir Crit Care Med 2020; 201:1050-1057. [PMID: 32176850 DOI: 10.1164/rccm.202002-0285up] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Yaseen M Arabi
- Intensive Care Department, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Rama Mallampalli
- Division of Pulmonary, Critical Care, and Sleep Medicine, Ohio State Wexner Medical, Center, Columbus, Ohio; and
| | - Joshua A Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, Ohio State Wexner Medical, Center, Columbus, Ohio; and
| | - Nicholas A Bosch
- Department of Medicine, Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Allan J Walkey
- Department of Medicine, Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Hasan M Al-Dorzi
- Intensive Care Department, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
50
|
Abstract
For infectious-disease outbreaks, clinical solutions typically focus on efficient pathogen destruction. However, the COVID-19 pandemic provides a reminder that infectious diseases are complex, multisystem conditions, and a holistic understanding will be necessary to maximize survival. For COVID-19 and all other infectious diseases, metabolic processes are intimately connected to the mechanisms of disease pathogenesis and the resulting pathology and pathophysiology, as well as the host defence response to the infection. Here, I examine the relationship between metabolism and COVID-19. I discuss why preexisting metabolic abnormalities, such as type 2 diabetes and hypertension, may be important risk factors for severe and critical cases of infection, highlighting parallels between the pathophysiology of these metabolic abnormalities and the disease course of COVID-19. I also discuss how metabolism at the cellular, tissue and organ levels might be harnessed to promote defence against the infection, with a focus on disease-tolerance mechanisms, and speculate on the long-term metabolic consequences for survivors of COVID-19.
Collapse
Affiliation(s)
- Janelle S Ayres
- Molecular and Systems Physiology Laboratory, Gene Expression Laboratory, NOMIS Center for Immunology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|