1
|
H Al-Qaisi TS, Jabbar AAJ, Raouf MMHM, AbdulSamad Ismail P, Mothana RA, Hawwal MF, Hassan RR, Abdulla MA, Saleh MI, Awad M. Persimmon (Diospyros kaki L.) leaves accelerates skin tissue regeneration in excisional wound model: possible molecular mechanisms. J Mol Histol 2025; 56:73. [PMID: 39849207 DOI: 10.1007/s10735-024-10304-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/09/2024] [Indexed: 01/25/2025]
Abstract
Persimmon (Diospyros kaki L.) leaves are a traditional medicinal herb used for treating many infectious and inflammatory-related conditions, including wound healing. To validate its traditional use, our study evaluates the acute toxicity and wound-healing effects of methanolic extracts of Persimmon (Diospyros kaki L.) leaves (MEPL) on excisional neck injury in rats. A uniform dorsal neck injury was created for twenty-four Sprague Dawley rats, which were randomly aligned into 4 groups and treated topically twice daily with 0.2 ml of the following: group A, rats treated with 1% CMC; group B, rats received intrasite gel; groups C and D, rats treated with MEPL (0.2 ml of 250 and 500 mg/kg, respectively). The toxicity results showed a lack of physiologic alteration or mortality in rats ingested with an oral dosage of up to 5 g/kg of MEPL. Histological screening of regenerated skin tissues revealed higher deposition of collagen, fibroblast cells, and reduced inflammatory cells in MEPL-treated rats. The topical application of MEPL led to positive modulation of Transforming Growth Factor Beta 1 (angiogenetic factor) in wound tissues, indicating increased tissue regeneration and faster wound contraction. MEPL treatment caused a significant elevation of tissue antioxidants (superoxide dismutase and catalase) and hydroxyproline (collagen) contents while reducing malondialdehyde contents. The inflammatory mediators (TNF-α and IL-6) were lower, and anti-inflammatory cytokines (interleukin 10) were higher in MEPL-treated rats than in the vehicle group. The study outcomes back up the traditional use of MEPL for wound healing, which could be linked with its phytochemicals (flavonoids and terpenoids) that require further isolation and molecular identification.
Collapse
Affiliation(s)
- Talal Salem H Al-Qaisi
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, P.O. Box 59911, Abu Dhabi, United Arab Emirates
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Ahmed A J Jabbar
- Department of Medical Laboratory Technology, Erbil Technical Health and Medical College, Erbil Polytechnic University, Erbil, 44001, Iraq.
| | - Mohammed M Hussein M Raouf
- Department of Biomedical Sciences, College of Applied Science, Cihan University-Erbil, Kurdistan Region, Erbil, 44001, Iraq
| | | | - Ramzi A Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Mohammed F Hawwal
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Rawaz Rizgar Hassan
- Department of Medical microbiology, College of Science, Knowledge University, Kirkuk Road, Erbil, 44001, Iraq
| | - Mahmood Ameen Abdulla
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Musher Ismael Saleh
- Department of Chemistry, Faculty of Science and Health, Koya University, Koya KOY45, Kurdistan Region, Erbil, 44001, Iraq
| | - Mohammed Awad
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, ON, Canada
- School of Engineering, Samarkand International University of Technology, 270 Spitamen Ave, Samarkand, 140100, Uzbekistan
| |
Collapse
|
2
|
Zhou C, Bao G, Chen Y. TRIM46 accelerates H1N1 influenza virus-induced ferroptosis and inflammatory response by regulating SLC7A11 ubiquitination. J Bioenerg Biomembr 2024; 56:631-643. [PMID: 39531094 DOI: 10.1007/s10863-024-10043-w] [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: 07/06/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Influenza A (H1N1) virus is an acute respiratory infection responsible for enormous morbidity and mortality worldwide. The tripartite motif-containing protein 46 (TRIM46) has an antiviral function that inhibits various viral infections. This study is designed to explore the role and mechanism of TRIM46 in the progress of H1N1 infection. Herein, we infected A549 or 16HBE cells with the H1N1 virus at different times to assess TRIM46 and solute carrier family 7 member 11 (SLC7A11) expression. TRIM46 and Influenza A nucleoprotein mRNA levels were detected by real-time quantitative polymerase chain reaction (RT-qPCR). TRIM46, solute carrier family 7 member 11 (SLC7A11), and Nucleoprotein protein levels were detected using protein level were detected by western blot assay. Cell virulence was determined using Virulence assay (TCID50) assay. Cell viability was determined using Cell Counting Kit-8 (CCK-8) assay. Reactive oxygen species (ROS), intracellular iron content, Malondialdehyde (MDA), and Glutathione (GSH) levels were determined using special assay kits. The stability of SLC7A11 was assessed by Cycloheximide (CHX) assay. Interaction between TRIM46 and SLC7A11 was verified using Co-immunoprecipitation (CoIP) assay. The biological role of TRIM46 was assessed in H1N1 virus-challenged lung injury mice in vivo. TRIM46 level was significantly increased during H1N1 virus infection, and SLC7A11 expression was decreased. TRIM46 downregulation could suppress H1N1 virus replication and relieve H1N1 infection-induced ferroptosis and inflammation in A549 or 16HBE cells. Mechanistically, TRIM46 could promote SLC7A11 ubiquitination and decrease its stability. TRIM46 knockdown repressed H1N1 virus-induced lung injury in vivo. TRIM46 could contribute to influenza A H1N1 virus infection by promoting SLC7A11 ubiquitination in A549 cells, which indicates that targeting TRIM46 may improve the prognosis of patients.
Collapse
Affiliation(s)
- Chao Zhou
- Department of Infection, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang, China
| | - Genchong Bao
- Department of Neurology, The People's Hospital of Danyang, Danyang, China
| | - Yanfei Chen
- Department of ICU, Danyang Hospital of Traditional Chinese Medicine, No. 38, Yunyang Road, Danyang, 212300, China.
| |
Collapse
|
3
|
Sun ZC, Liao R, Xian C, Lin R, Wang L, Fang Y, Zhang Z, Liu Y, Wu J. Natural pachypodol integrated, lung targeted and inhaled lipid nanomedicine ameliorates acute lung injury via anti-inflammation and repairing lung barrier. J Control Release 2024; 375:300-315. [PMID: 39265826 DOI: 10.1016/j.jconrel.2024.09.013] [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: 05/15/2024] [Revised: 07/29/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Acute lung injury (ALI) or acute respiratory distress syndrome (ARDS) is a high-mortality disease caused by multiple disorders such as COVID-19, influenza, and sepsis. Current therapies mainly rely on the inhalation of nitric oxide or injection of pharmaceutical drugs (e.g., glucocorticoids); however, their toxicity, side effects, or administration routes limit their clinical application. In this study, pachypodol (Pac), a hydrophobic flavonol with anti-inflammatory effects, was extracted from Pogostemon cablin Benth and intercalated in liposomes (Pac@liposome, Pac-lipo) to improve its solubility, biodistribution, and bioavailability, aiming at enhanced ALI/ARDS therapy. Nanosized Pac-lipo was confirmed to have stable physical properties, good biodistribution, and reliable biocompatibility. In vitro tests proved that Pac-lipo has anti-inflammatory property and protective effects on endothelial and epithelial barriers in lipopolysaccharide (LPS)-induced macrophages and endothelial cells, respectively. Further, the roles of Pac-lipo were validated on treating LPS-induced ALI in mice. Pac-lipo showed better effects than did Pac alone on relieving ALI phenotypes: It significantly attenuated lung index, improved pulmonary functions, inhibited cytokine expression such as TNF-α, IL-6, IL-1β, and iNOS in lung tissues, alleviated lung injury shown by HE staining, reduced protein content and total cell number in bronchoalveolar lavage fluid, and repaired lung epithelial and vascular endothelial barriers. As regards the underlying mechanisms, RNA sequencing results showed that the effects of the drugs were associated with numerous immune- and inflammation-related signaling pathways. Molecular docking and western blotting demonstrated that Pac-lipo inhibited the activation of the TLR4-MyD88-NF-κB/MAPK signaling pathway. Taken together, for the first time, our new drug (Pac-lipo) ameliorates ALI via inhibition of TLR4-MyD88-NF-κB/MAPK pathway-mediated inflammation and disruption of lung barrier. These findings may provide a promising strategy for ALI treatment in the clinic.
Collapse
Affiliation(s)
- Zhi-Chao Sun
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China
| | - Ran Liao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China
| | - Caihong Xian
- Bioscience and Biomedical Engineering Thrust, Systems Hub, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, Guangdong, China; School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Ran Lin
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China
| | - Liying Wang
- Bioscience and Biomedical Engineering Thrust, Systems Hub, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, Guangdong, China; School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Yifei Fang
- Bioscience and Biomedical Engineering Thrust, Systems Hub, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, Guangdong, China; School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Zhongde Zhang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China.
| | - Yuntao Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, 55 N, Neihuanxi Road, Guangzhou 510006, Guangdong, China.
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, Systems Hub, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou 511400, Guangdong, China; Division of Life Science, The Hong Kong University of Science and Technology, 999077, Hong Kong SAR.
| |
Collapse
|
4
|
Wen Y, Liu Y, Liu W, Liu W, Dong J, Liu Q, Yu Z, Ren H, Hao H. Ferroptosis: a potential target for acute lung injury. Inflamm Res 2024; 73:1615-1629. [PMID: 39152299 DOI: 10.1007/s00011-024-01919-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/09/2024] [Accepted: 07/10/2024] [Indexed: 08/19/2024] Open
Abstract
Acute lung injury (ALI) is caused by a variety of intrapulmonary and extrapulmonary factors and is associated with high morbidity and mortality. Oxidative stress is an important part of the pathological mechanism of ALI. Ferroptosis is a mode of programmed cell death distinguished from others and characterized by iron-dependent lipid peroxidation. This article reviews the metabolic regulation of ferroptosis, its role in the pathogenesis of ALI, and the use of ferroptosis as a therapeutic target regarding the pharmacological treatment of ALI.
Collapse
Affiliation(s)
- Yuqi Wen
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Yang Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Weihong Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Wenli Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Jinyan Dong
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Qingkuo Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Zhen Yu
- Jinan Family Planning Service Center, Jinan, 250014, China
| | - Hongsheng Ren
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250014, China.
| | - Hao Hao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| |
Collapse
|
5
|
Xiong L, Liu Y, Wang Y, Zhao H, Song X, Fan W, Zhang L, Zhang Y. The protective effect of Lonicera japonica Thunb. against lipopolysaccharide-induced acute lung injury in mice: Modulation of inflammation, oxidative stress, and ferroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118333. [PMID: 38750986 DOI: 10.1016/j.jep.2024.118333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Various components of Lonicera japonica Thunb. (LJT) exhibit pharmacological activities, including anti-inflammatory and antioxidant effects. Nevertheless, the relationship between LJT and ferroptosis remains largely unexplored. AIM OF THE STUDY The purpose of this research was to look into the role of LJT in regulating LPS-induced ferroptosis in ALI and to compare the effects of different parts of LJT. MATERIALS AND METHODS We established a mice ALI model by treating with LPS. Administered mice with different doses of Lonicerae Japonicae Flos (LJF), Lonicera Japonica Leaves (LJL) and Lonicerae Caulis (LRC) extracts, respectively. The levels of IL-6, IL-1β, TNF-α, IL-4, IL-10, and PGE2 in bronchoalveolar lavage fluid (BALF) were measured using enzyme-linked immunosorbent assay. Furthermore, the concentrations of superoxide dismutase (SOD), malondialdehyde (MDA), glutathione (GSH), reactive oxygen species (ROS), and total ferrous ions (Fe2+) in lung tissues were evaluated. Hematoxylin and eosin staining was conducted to examine the morphological structure of lung tissues. Transmission electron microscopy was used to investigate the ultrastructural morphology of mitochondria. Furthermore, the effects of LJT were evaluated via immunohistochemical staining, western blotting, and quantitative real-time polymerase chain reaction analyses. Finally, employing molecular docking and molecular dynamics research techniques, we aimed to identify crucial components in LJT that might inhibit ferroptosis by targeting nuclear factor erythroid 2-related factor 2 (Nrf2) and glutathione peroxidase 4 (GPX4). RESULTS We observed that pretreatment with LJT significantly mitigated LPS-induced lung injury and suppressed ferroptosis. This was supported by reduced accumulation of pro-inflammatory cytokines, ROS, MDA, and Fe2+, along with increased levels of anti-inflammatory cytokines, SOD, GSH, Nrf2, and GPX4 in the lung tissues of ALI mice. Luteolin-7-O-rutinoside, apigenin-7-O-rutinoside, and amentoflavone in LJT exhibit excellent docking effects with key targets of ferroptosis, Nrf2 and GPX4. CONCLUSIONS Pretreatment with LJT may alleviate LPS-induced ALI, possibly by suppressing ferroptosis. Our initial results indicate that LJT activates the Nrf2/GPX4 axis, providing protection against ferroptosis in ALI. This finding offers a promising therapeutic candidate for ALI treatment.
Collapse
Affiliation(s)
- Lewen Xiong
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yan Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Yang Wang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Hongwei Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xiaochen Song
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenjing Fan
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Longfei Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Yongqing Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
6
|
Letafati A, Taghiabadi Z, Ardekani OS, Abbasi S, Najafabadi AQ, Jazi NN, Soheili R, Rodrigo R, Yavarian J, Saso L. Unveiling the intersection: ferroptosis in influenza virus infection. Virol J 2024; 21:185. [PMID: 39135112 PMCID: PMC11321227 DOI: 10.1186/s12985-024-02462-3] [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: 05/30/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
The influenza virus (IFV) imposes a considerable health and economic burden globally, requiring a comprehensive understanding of its pathogenic mechanisms. Ferroptosis, an iron-dependent lipid peroxidation cell death pathway, holds unique implications for the antioxidant defense system, with possible contributions to inflammation. This exploration focuses on the dynamic interplay between ferroptosis and the host defense against viruses, emphasizing the influence of IFV infections on the activation of the ferroptosis pathway. IFV causes different types of cell death, including apoptosis, necrosis, and ferroptosis. IFV-induced ferroptotic cell death is mediated by alterations in iron homeostasis, intensifying the accumulation of reactive oxygen species and promoting lipid peroxidation. A comprehensive investigation into the mechanism of ferroptosis in viral infections, specifically IFV, has great potential to identify therapeutic strategies. This understanding may pave the way for the development of drugs using ferroptosis inhibitors, presenting an effective approach to suppress viral infections.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Taghiabadi
- Department of Microbiology and Virology of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Omid Salahi Ardekani
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Simin Abbasi
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Qaraee Najafabadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Negar Nayerain Jazi
- Department of Bacteriology & Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roben Soheili
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jila Yavarian
- Department of Virology, Faculty of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University, Rome, Italy.
| |
Collapse
|
7
|
Zheng J, Li Y, Kong X, Guo J. Exploring immune-related pathogenesis in lung injury: Providing new insights Into ALI/ARDS. Biomed Pharmacother 2024; 175:116773. [PMID: 38776679 DOI: 10.1016/j.biopha.2024.116773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/08/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) represent a significant global burden of morbidity and mortality, with lung injury being the primary cause of death in affected patients. The pathogenesis of lung injury, however, remains a complex issue. In recent years, the role of the immune system in lung injury has attracted extensive attention worldwide. Despite advancements in our understanding of various lung injury subtypes, significant limitations persist in both prevention and treatment. This review investigates the immunopathogenesis of ALI/ARDS, aiming to elucidate the pathological processes of lung injury mediated by dendritic cells (DCs), natural killer (NK) cells, phagocytes, and neutrophils. Furthermore, the article expounds on the critical contributions of gut microbiota, inflammatory pathways, and cytokine storms in the development of ALI/ARDS.
Collapse
Affiliation(s)
- Jiajing Zheng
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Li
- Pharmacy Department of the First Affiliated Hospital, Henan University of Science and Technology, Luoyang 471000, China
| | - Xianbin Kong
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Jinhe Guo
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
8
|
Chuang YT, Yen CY, Chien TM, Chang FR, Tsai YH, Wu KC, Tang JY, Chang HW. Ferroptosis-Regulated Natural Products and miRNAs and Their Potential Targeting to Ferroptosis and Exosome Biogenesis. Int J Mol Sci 2024; 25:6083. [PMID: 38892270 PMCID: PMC11173094 DOI: 10.3390/ijms25116083] [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: 04/28/2024] [Revised: 05/22/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Ferroptosis, which comprises iron-dependent cell death, is crucial in cancer and non-cancer treatments. Exosomes, the extracellular vesicles, may deliver biomolecules to regulate disease progression. The interplay between ferroptosis and exosomes may modulate cancer development but is rarely investigated in natural product treatments and their modulating miRNAs. This review focuses on the ferroptosis-modulating effects of natural products and miRNAs concerning their participation in ferroptosis and exosome biogenesis (secretion and assembly)-related targets in cancer and non-cancer cells. Natural products and miRNAs with ferroptosis-modulating effects were retrieved and organized. Next, a literature search established the connection of a panel of ferroptosis-modulating genes to these ferroptosis-associated natural products. Moreover, ferroptosis-associated miRNAs were inputted into the miRNA database (miRDB) to bioinformatically search the potential targets for the modulation of ferroptosis and exosome biogenesis. Finally, the literature search provided a connection between ferroptosis-modulating miRNAs and natural products. Consequently, the connections from ferroptosis-miRNA-exosome biogenesis to natural product-based anticancer treatments are well-organized. This review sheds light on the research directions for integrating miRNAs and exosome biogenesis into the ferroptosis-modulating therapeutic effects of natural products on cancer and non-cancer diseases.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan
| | - Tsu-Ming Chien
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan;
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Urology, Kaohsiung Gangshan Hospital, Kaohsiung Medical University, Kaohsiung 820111, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung 907101, Taiwan;
| | - Kuo-Chuan Wu
- Department of Computer Science and Information Engineering, National Pingtung University, Pingtung 900391, Taiwan;
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| |
Collapse
|
9
|
Jia KW, Yao RQ, Fan YW, Zhang DJ, Zhou Y, Wang MJ, Zhang LY, Dong Y, Li ZX, Wang SY, Wang M, Li YH, Zhang LX, Lei T, Gui LC, Lu S, Yang YY, Wang SX, Yu YZ, Yao YM, Hou J. Interferon-α stimulates DExH-box helicase 58 to prevent hepatocyte ferroptosis. Mil Med Res 2024; 11:22. [PMID: 38622688 PMCID: PMC11017495 DOI: 10.1186/s40779-024-00524-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 03/19/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Liver ischemia/reperfusion (I/R) injury is usually caused by hepatic inflow occlusion during liver surgery, and is frequently observed during war wounds and trauma. Hepatocyte ferroptosis plays a critical role in liver I/R injury, however, it remains unclear whether this process is controlled or regulated by members of the DEAD/DExH-box helicase (DDX/DHX) family. METHODS The expression of DDX/DHX family members during liver I/R injury was screened using transcriptome analysis. Hepatocyte-specific Dhx58 knockout mice were constructed, and a partial liver I/R operation was performed. Single-cell RNA sequencing (scRNA-seq) in the liver post I/R suggested enhanced ferroptosis by Dhx58hep-/-. The mRNAs and proteins associated with DExH-box helicase 58 (DHX58) were screened using RNA immunoprecipitation-sequencing (RIP-seq) and IP-mass spectrometry (IP-MS). RESULTS Excessive production of reactive oxygen species (ROS) decreased the expression of the IFN-stimulated gene Dhx58 in hepatocytes and promoted hepatic ferroptosis, while treatment using IFN-α increased DHX58 expression and prevented ferroptosis during liver I/R injury. Mechanistically, DHX58 with RNA-binding activity constitutively associates with the mRNA of glutathione peroxidase 4 (GPX4), a central ferroptosis suppressor, and recruits the m6A reader YT521-B homology domain containing 2 (YTHDC2) to promote the translation of Gpx4 mRNA in an m6A-dependent manner, thus enhancing GPX4 protein levels and preventing hepatic ferroptosis. CONCLUSIONS This study provides mechanistic evidence that IFN-α stimulates DHX58 to promote the translation of m6A-modified Gpx4 mRNA, suggesting the potential clinical application of IFN-α in the prevention of hepatic ferroptosis during liver I/R injury.
Collapse
Affiliation(s)
- Kai-Wei Jia
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ren-Qi Yao
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100853, China
| | - Yi-Wen Fan
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ding-Ji Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ye Zhou
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Min-Jun Wang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Li-Yuan Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Yue Dong
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Zhi-Xuan Li
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Su-Yuan Wang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Mu Wang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Yun-Hui Li
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Lu-Xin Zhang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ting Lei
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Liang-Chen Gui
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Shan Lu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Ying-Yun Yang
- Center for Immunotherapy, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Si-Xian Wang
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Yi-Zhi Yu
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing, 100853, China.
| | - Jin Hou
- National Key Laboratory of Medical Immunology & Institute of Immunology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
10
|
Chen T, Ding L, Zhao M, Song S, Hou J, Li X, Li M, Yin K, Li X, Wang Z. Recent advances in the potential effects of natural products from traditional Chinese medicine against respiratory diseases targeting ferroptosis. Chin Med 2024; 19:49. [PMID: 38519984 PMCID: PMC10958864 DOI: 10.1186/s13020-024-00918-w] [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: 10/12/2023] [Accepted: 03/07/2024] [Indexed: 03/25/2024] Open
Abstract
Respiratory diseases, marked by structural changes in the airways and lung tissues, can lead to reduced respiratory function and, in severe cases, respiratory failure. The side effects of current treatments, such as hormone therapy, drugs, and radiotherapy, highlight the need for new therapeutic strategies. Traditional Chinese Medicine (TCM) offers a promising alternative, leveraging its ability to target multiple pathways and mechanisms. Active compounds from Chinese herbs and other natural sources exhibit anti-inflammatory, antioxidant, antitumor, and immunomodulatory effects, making them valuable in preventing and treating respiratory conditions. Ferroptosis, a unique form of programmed cell death (PCD) distinct from apoptosis, necrosis, and others, has emerged as a key area of interest. However, comprehensive reviews on how natural products influence ferroptosis in respiratory diseases are lacking. This review will explore the therapeutic potential and mechanisms of natural products from TCM in modulating ferroptosis for respiratory diseases like acute lung injury (ALI), asthma, pulmonary fibrosis (PF), chronic obstructive pulmonary disease (COPD), lung ischemia-reperfusion injury (LIRI), pulmonary hypertension (PH), and lung cancer, aiming to provide new insights for research and clinical application in TCM for respiratory health.
Collapse
Affiliation(s)
- Tian Chen
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Lu Ding
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
- Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130021, China
| | - Meiru Zhao
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Siyu Song
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Juan Hou
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xueyan Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Min Li
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Kai Yin
- College of Integrated Traditional Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China.
| | - Zeyu Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China.
| |
Collapse
|
11
|
Zhang F, Xiang Y, Ma Q, Guo E, Zeng X. A deep insight into ferroptosis in lung disease: facts and perspectives. Front Oncol 2024; 14:1354859. [PMID: 38562175 PMCID: PMC10982415 DOI: 10.3389/fonc.2024.1354859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
In the last decade, ferroptosis has received much attention from the scientific research community. It differs from other modes of cell death at the morphological, biochemical, and genetic levels. Ferroptosis is mainly characterized by non-apoptotic iron-dependent cell death caused by iron-dependent lipid peroxide excess and is accompanied by abnormal iron metabolism and oxidative stress. In recent years, more and more studies have shown that ferroptosis is closely related to the occurrence and development of lung diseases. COPD, asthma, lung injury, lung fibrosis, lung cancer, lung infection and other respiratory diseases have become the third most common chronic diseases worldwide, bringing serious economic and psychological burden to people around the world. However, the exact mechanism by which ferroptosis is involved in the development and progression of lung diseases has not been fully revealed. In this manuscript, we describe the mechanism of ferroptosis, targeting of ferroptosis related signaling pathways and proteins, summarize the relationship between ferroptosis and respiratory diseases, and explore the intervention and targeted therapy of ferroptosis for respiratory diseases.
Collapse
Affiliation(s)
- Fan Zhang
- Wuhan University of Science and Technology, School of Medicine, Wuhan, China
| | - Yu Xiang
- Wuhan University of Science and Technology, School of Medicine, Wuhan, China
| | - Qiao Ma
- Wuhan University of Science and Technology, School of Medicine, Wuhan, China
| | - E. Guo
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Xiansheng Zeng
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
12
|
Feng S, Tang D, Wang Y, Li X, Bao H, Tang C, Dong X, Li X, Yang Q, Yan Y, Yin Z, Shang T, Zheng K, Huang X, Wei Z, Wang K, Qi S. The mechanism of ferroptosis and its related diseases. MOLECULAR BIOMEDICINE 2023; 4:33. [PMID: 37840106 PMCID: PMC10577123 DOI: 10.1186/s43556-023-00142-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 10/17/2023] Open
Abstract
Ferroptosis, a regulated form of cellular death characterized by the iron-mediated accumulation of lipid peroxides, provides a novel avenue for delving into the intersection of cellular metabolism, oxidative stress, and disease pathology. We have witnessed a mounting fascination with ferroptosis, attributed to its pivotal roles across diverse physiological and pathological conditions including developmental processes, metabolic dynamics, oncogenic pathways, neurodegenerative cascades, and traumatic tissue injuries. By unraveling the intricate underpinnings of the molecular machinery, pivotal contributors, intricate signaling conduits, and regulatory networks governing ferroptosis, researchers aim to bridge the gap between the intricacies of this unique mode of cellular death and its multifaceted implications for health and disease. In light of the rapidly advancing landscape of ferroptosis research, we present a comprehensive review aiming at the extensive implications of ferroptosis in the origins and progress of human diseases. This review concludes with a careful analysis of potential treatment approaches carefully designed to either inhibit or promote ferroptosis. Additionally, we have succinctly summarized the potential therapeutic targets and compounds that hold promise in targeting ferroptosis within various diseases. This pivotal facet underscores the burgeoning possibilities for manipulating ferroptosis as a therapeutic strategy. In summary, this review enriched the insights of both investigators and practitioners, while fostering an elevated comprehension of ferroptosis and its latent translational utilities. By revealing the basic processes and investigating treatment possibilities, this review provides a crucial resource for scientists and medical practitioners, aiding in a deep understanding of ferroptosis and its effects in various disease situations.
Collapse
Affiliation(s)
- Shijian Feng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Dan Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yichang Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiang Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Hui Bao
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Chengbing Tang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiuju Dong
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xinna Li
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinxue Yang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yun Yan
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhijie Yin
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tiantian Shang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Kaixuan Zheng
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaofang Huang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zuheng Wei
- Chengdu Jinjiang Jiaxiang Foreign Languages High School, Chengdu, People's Republic of China
| | - Kunjie Wang
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Shiqian Qi
- Department of Urology and Institute of Urology (Laboratory of Reconstructive Urology), State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
13
|
Huang J, Ma X, Liao Z, Liu Z, Wang K, Feng Z, Ning Y, Lu F, Li L. Network pharmacology and experimental validation of Maxing Shigan decoction in the treatment of influenza virus-induced ferroptosis. Chin J Nat Med 2023; 21:775-788. [PMID: 37879795 DOI: 10.1016/s1875-5364(23)60457-1] [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: 04/16/2023] [Indexed: 10/27/2023]
Abstract
Influenza is an acute viral respiratory infection that has caused high morbidity and mortality worldwide. Influenza A virus (IAV) has been found to activate multiple programmed cell death pathways, including ferroptosis. Ferroptosis is a novel form of programmed cell death in which the accumulation of intracellular iron promotes lipid peroxidation, leading to cell death. However, little is known about how influenza viruses induce ferroptosis in the host cells. In this study, based on network pharmacology, we predicted the mechanism of action of Maxing Shigan decoction (MXSGD) in IAV-induced ferroptosis, and found that this process was related to biological processes, cellular components, molecular function and multiple signaling pathways, where the hypoxia inducible factor-1(HIF-1) signaling pathway plays a significant role. Subsequently, we constructed the mouse lung epithelial (MLE-12) cell model by IAV-infected in vitro cell experiments, and revealed that IAV infection induced cellular ferroptosis that was characterized by mitochondrial damage, increased reactive oxygen species (ROS) release, increased total iron and iron ion contents, decreased expression of ferroptosis marker gene recombinant glutathione peroxidase 4 (GPX4), increased expression of acyl-CoA synthetase long chain family member 4 (ACSL4), and enhanced activation of hypoxia inducible factor-1α (HIF-1α), induced nitric oxide synthase (iNOS) and vascular endothelial growth factor (VEGF) in the HIF-1 signaling pathway. Treatment with MXSGD effectively reduced intracellular viral load, while reducing ROS, total iron and ferrous ion contents, repairing mitochondrial results and inhibiting the expression of cellular ferroptosis and the HIF-1 signaling pathway. Finally, based on animal experiments, it was found that MXSGD effectively alleviated pulmonary congestion, edema and inflammation in IAV-infected mice, and inhibited the expression of ferroptosis-related protein and the HIF-1 signaling pathway in lung tissues.
Collapse
Affiliation(s)
- Jiawang Huang
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xinyue Ma
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zexuan Liao
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhuolin Liu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Kangyu Wang
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Zhiying Feng
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yi Ning
- The Medicine School of Hunan University of Chinese Medicine, Changsha 410208, China
| | - Fangguo Lu
- The Medicine School of Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ling Li
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China; Hunan Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha 410208, China.
| |
Collapse
|
14
|
Ohno M, Gowda SGB, Sekiya T, Nomura N, Shingai M, Hui SP, Kida H. The elucidation of plasma lipidome profiles during severe influenza in a mouse model. Sci Rep 2023; 13:14210. [PMID: 37648726 PMCID: PMC10469212 DOI: 10.1038/s41598-023-41055-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: 04/04/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
Although influenza virus infection has been shown to affect lipid metabolism, details remain unknown. Therefore, we elucidated the kinetic lipid profiles of mice infected with different doses of influenza virus A/Puerto Rico/8/34 (H1N1) (PR8) by measuring multiple lipid molecular species using untargeted lipidomic analysis. C57BL/6 male mice were intranasally infected with PR8 virus at 50 or 500 plaque-forming units to cause sublethal or lethal influenza, respectively. Plasma and tissue samples were collected at 1, 3, and 6 days post-infection (dpi), and comprehensive lipidomic analysis was performed using high-performance liquid chromatography-linear trap quadrupole-Orbitrap mass spectrometry, as well as gene expression analyses. The most prominent feature of the lipid profile in lethally infected mice was the elevated plasma concentrations of phosphatidylethanolamines (PEs) containing polyunsaturated fatty acid (PUFA) at 3 dpi. Furthermore, the facilitation of PUFA-containing phospholipid production in the lungs, but not in the liver, was suggested by gene expression and lipidomic analysis of tissue samples. Given the increased plasma or serum levels of PUFA-containing PEs in patients with other viral infections, especially in severe cases, the elevation of these phospholipids in circulation could be a biomarker of infection and the severity of infectious diseases.
Collapse
Affiliation(s)
- Marumi Ohno
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
| | - Siddabasave Gowda B Gowda
- Faculty of Health Sciences, Hokkaido University, Kita-12 Nishi-5, Kita-ku, Sapporo, 060-0812, Japan
- Graduate School of Global Food Resources, Hokkaido University, Sapporo, Japan
| | - Toshiki Sekiya
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Naoki Nomura
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
| | - Masashi Shingai
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Kita-12 Nishi-5, Kita-ku, Sapporo, 060-0812, Japan.
| | - Hiroshi Kida
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Kita 20 Nishi 10, Kita-ku, Sapporo, 001-0020, Japan.
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan.
| |
Collapse
|
15
|
Park EJ, Jin SW, Shim I, Cho AE. Comparison of disinfectants-induced gene expression profile: Potential adverse effects. Toxicol Appl Pharmacol 2023; 470:116546. [PMID: 37149095 PMCID: PMC10158043 DOI: 10.1016/j.taap.2023.116546] [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] [Received: 03/20/2023] [Revised: 04/29/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
Despite their importance in combating the spread of the COVID-19 pandemic, adverse effects of disinfectants on human health, especially the respiratory system, have been of continuing concern to researchers. Considering that bronchi are the main target of sprayed disinfectants, we here treated the seven major active ingredients in disinfectant products accepted by the US EPA to human bronchial epithelial cells and determined the subtoxic levels. Then, we performed microarray analysis using total RNA obtained at the subtoxic level and designed a network representing disinfectant-induced cellular response using the KEGG pathway analysis technique. Polyhexamethylguanidine phosphate, a lung fibrosis inducer, was used as a reference material to verify the relationship between cell death and pathology. The derived results reveal potential adverse effects along with the need for an effective application strategy for each chemical.
Collapse
Affiliation(s)
- Eun-Jung Park
- Graduate School of Medicine, Kyung Hee University, 02447, Republic of Korea; Human Health and Environmental Toxins Research Center, Kyung Hee University, 02447, Republic of Korea.
| | - Seung-Woo Jin
- Department of Biomedical Science and Technology, Graduate school, 02447 Kyung Hee University, Republic of Korea
| | - Ilseob Shim
- Environmental Health Research Department, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Art E Cho
- Department of Biotechnology and Bioinformatics, Korea University, Jochiwon, Chungnam 339-700, Republic of Korea
| |
Collapse
|