1
|
Shi Y, Yang X, Jiang H, Wu S, Hong Y, Su W, Wang X. Alpinia officinarum Hance extract relieved sepsis-induced myocardial ferroptosis and inflammation by inhibiting lncRNA MIAT/TRAF6/NF-κB axis. Allergol Immunopathol (Madr) 2024; 52:21-28. [PMID: 39278847 DOI: 10.15586/aei.v52i5.1035] [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: 07/05/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024]
Abstract
Sepsis is generally triggered by a dysfunctional host response to infection, and it can result in life-threatening organ dysfunction. Alpinia officinarum Hance (AO) exhibits regulatory functions in some diseases. However, whether AO extract (AOE) plays a promoting role in sepsis--triggered myocardial injury is unclear. This study was aimed at investigating the regulatory effects of AOE on myocardial ferroptosis and inflammation in sepsis, and the regulation effects on the lncRNA MIAT/TRAF6/NF-κB axis. Lipopolysaccharide (LPS) was used to treat mice for establishing an in vivo sepsis model. The pathological changes in heart tissues were observed through hematoxylin-eosin (HE) staining. The levels of CK-MB, cTnl, MDA, SOD, IL-1β, IL-18, IL-6, and TNF-α in serum were detected through enzyme-linked immunosorbent assay (ELISA). The level of Fe2+ was assessed, and the protein expressions (ACSL4, GPX4, TRAF6, p-P65, and P65) were examined through western blot. The expressions of lncRNA MIAT and TRAF6 were measured through real-time quantitative polymerase chain reaction (RT-qPCR). Our results demonstrated that AOE treatment ameliorated sepsis-triggered myocardial damage by reducing the disordered cardiomyocytes, the destroyed sarcolemma, and the CK-MB and cTnl levels. In addition, AOE treatment inhibited sepsis-induced myocardial ferroptosis and inflammation by regulating Fe2+, ACSL4, GPX4, IL-1β, IL-18, IL-6, and TNF-α levels. Moreover, the improvement effect of AOE was strengthened with the increase in the dose of AOE (25, 50, 100 mg/kg). It was also revealed that AOE treatment retarded the lncRNA MIAT/TRAF6/NF-κB axis. Rescue assays manifested that overexpression of MIAT reduced the cardioprotective effect of AOE. In conclusion, AOE relieved sepsis-induced myocardial ferroptosis and inflammation by inhibiting lncRNA MIAT/TRAF6/NF-κB axis. These findings may provide a potential therapeutic drug for the treatment of sepsis.
Collapse
Affiliation(s)
- Yao Shi
- Department of Pediatrics, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobo Yang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China;
| | - Hong Jiang
- Department of Pediatrics, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanxia Wu
- Department of Pediatrics, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Hong
- Department of Pediatrics, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Su
- Department of Pediatrics, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuan Wang
- Department of Pediatrics, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Xie L, Zhang G, Wu Y, Hua Y, Ding W, Han X, Liu B, Zhou C, Li A. Protective effects of Wenqingyin on sepsis-induced acute lung injury through regulation of the receptor for advanced glycation end products pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155654. [PMID: 38723525 DOI: 10.1016/j.phymed.2024.155654] [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: 10/08/2023] [Revised: 02/06/2024] [Accepted: 04/17/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Wenqingyin (WQY), an ancient Chinese medicinal agent, has been extensively used in treating infectious ailments throughout history. However, the anti-sepsis mechanism remains unknown. PURPOSE This study investigated the diverse mechanisms of WQY in mitigating sepsis-induced acute lung injury (ALI). Additionally, the effects of WQY were validated using biological experiments. METHODS This study combined UHPLC-Orbitrap-HRMS analysis and network pharmacology to predict the potential anti-sepsis mechanism of WQY. Sepsis-induced ALI models were established in vivo via intraperitoneal lipopolysaccharide (LPS) administration and in vitro by LPS-stimulated RAW 264.7 macrophages. Various techniques, including hematoxylin-eosin staining, TUNEL, qPCR, and ELISA, were used to assess lung damage and quantify inflammatory cytokines. Inflammatory cell infiltration was visualized through immunohistochemistry. Hub targets and signaling pathways were identified using Western blotting, immunohistochemistry, and immunofluorescence staining. RESULTS Seventy-five active components and 237 associated targets were acquired, with 145 of these targets overlapping with processes related to sepsis. Based on the comprehensive protein-protein interaction network analysis, JUN, AKT1, TP53, IL-6, HSP90AA1, CASP3, VEGFA, IL-1β, RELA, and EGFR may be targets of WQY for sepsis. Analysis of the Kyoto Gene and Genome Encyclopedia revealed that WQY is implicated in the advanced glycation end products/receptor for advanced glycation end products (AGE/RAGE) signaling pathway. In vivo, WQY alleviated sepsis-induced ALI, suppressing proinflammatory cytokines and inhibiting macrophage/neutrophil infiltration. In vitro, WQY reduced TNF-α, IL-6, and IL-1β in LPS-induced RAW 264.7 macrophages. Furthermore, we verified that WQY protected against sepsis-induced ALI by regulating the RAGE pathway for the first time. Baicalin, coptisine, and paeoniflorin may be the effective components of WQY that inhibit RAGE. CONCLUSION The primary mechanism of WQY in combating sepsis-induced ALI involves controlling RAGE levels and the PI3K/AKT pathway, suppressing inflammation, and mitigating lung damage. This study establishes a scientific foundation for understanding the mechanism of WQY and its clinical use in treating sepsis.
Collapse
Affiliation(s)
- Lingpeng Xie
- Department of Hepatology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510315, China
| | - Guoyong Zhang
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuting Wu
- Binzhou Medical University Hospital, Binzhou 256603, China
| | - Yue Hua
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Wenjun Ding
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xin Han
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China.
| | - Chuying Zhou
- Department of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Aimin Li
- Department of Hepatology, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou 510315, China.
| |
Collapse
|
3
|
Zhu M, Zhao T, Zha B, Zhang G, Qian W, Wang X, Zhao Q, Chen S, Hu Z, Dong L. Piceatannol protects against myocardial ischemia/reperfusion injury by inhibiting ferroptosis via Nrf-2 signaling-mediated iron metabolism. Biochem Biophys Res Commun 2024; 700:149598. [PMID: 38308910 DOI: 10.1016/j.bbrc.2024.149598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Myocardial tissue ischemia damages myocardial cells. Although reperfusion is an effective technique to rescue myocardial cell damage, it may also exacerbate myocardial cell damage. Ferroptosis, an iron-dependent cell death, occurs following myocardial ischemia-reperfusion (I/R). Piceatannol (PCT) is a natural stilbene compound with excellent antioxidant properties that protect against I/R injury and exerts protective effects against ferroptosis-induced cardiomyocytes following I/R injury; however, the exact mechanism remains to be elucidated. PURPOSE This study aims to investigate the protective effect and mechanism of PCT on myocardial ischemia-reperfusion injury. METHODS An ischemia-reperfusion model was established via ligation of the left anterior descending branch of mice's hearts and hypoxia-reoxygenation (H/R) of cardiomyocytes. RESULTS During ischemia-reperfusion, Nuclear factor E2-related factor 2 (Nrf-2) expression was downregulated, the left ventricular function was impaired, intracellular iron and lipid peroxidation product levels were elevated, and cardiomyocytes underwent ferroptosis. Furthermore, ferroptosis was enhanced following treatment with an Nrf-2 inhibitor. After PCT treatment, Nrf-2 expression significantly increased, intracellular ferrous ions and lipid peroxidation products significantly reduced, Ferroportin1 (FPN1) expression increased, and transferrin receptor-1 (TfR-1) expression was inhibited. CONCLUSIONS PCT regulates iron metabolism through Nrf-2 to protect against myocardial cell ferroptosis induced by myocardial I/R injury.
Collapse
Affiliation(s)
- Mengmei Zhu
- Department of Pharmacology, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Tianhao Zhao
- Department of Pharmacology, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Binshan Zha
- Department of Vascular and Thyroid Surgery, Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230031, China
| | - Guiyang Zhang
- Department of Pharmacology, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Weiwei Qian
- Department of Pharmacology, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Xinya Wang
- Department of Pharmacology, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Qiuju Zhao
- Department of Pharmacology, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Shuo Chen
- Department of Pharmacology, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Zeping Hu
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230031, China
| | - Liuyi Dong
- Department of Pharmacology, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, 230032, China.
| |
Collapse
|
4
|
Jiang H, Yang J, Li T, Wang X, Fan Z, Ye Q, Du Y. JAK/STAT3 signaling in cardiac fibrosis: a promising therapeutic target. Front Pharmacol 2024; 15:1336102. [PMID: 38495094 PMCID: PMC10940489 DOI: 10.3389/fphar.2024.1336102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/18/2024] [Indexed: 03/19/2024] Open
Abstract
Cardiac fibrosis is a serious health problem because it is a common pathological change in almost all forms of cardiovascular diseases. Cardiac fibrosis is characterized by the transdifferentiation of cardiac fibroblasts (CFs) into cardiac myofibroblasts and the excessive deposition of extracellular matrix (ECM) components produced by activated myofibroblasts, which leads to fibrotic scar formation and subsequent cardiac dysfunction. However, there are currently few effective therapeutic strategies protecting against fibrogenesis. This lack is largely because the molecular mechanisms of cardiac fibrosis remain unclear despite extensive research. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling cascade is an extensively present intracellular signal transduction pathway and can regulate a wide range of biological processes, including cell proliferation, migration, differentiation, apoptosis, and immune response. Various upstream mediators such as cytokines, growth factors and hormones can initiate signal transmission via this pathway and play corresponding regulatory roles. STAT3 is a crucial player of the JAK/STAT pathway and its activation is related to inflammation, malignant tumors and autoimmune illnesses. Recently, the JAK/STAT3 signaling has been in the spotlight for its role in the occurrence and development of cardiac fibrosis and its activation can promote the proliferation and activation of CFs and the production of ECM proteins, thus leading to cardiac fibrosis. In this manuscript, we discuss the structure, transactivation and regulation of the JAK/STAT3 signaling pathway and review recent progress on the role of this pathway in cardiac fibrosis. Moreover, we summarize the current challenges and opportunities of targeting the JAK/STAT3 signaling for the treatment of fibrosis. In summary, the information presented in this article is critical for comprehending the role of the JAK/STAT3 pathway in cardiac fibrosis, and will also contribute to future research aimed at the development of effective anti-fibrotic therapeutic strategies targeting the JAK/STAT3 signaling.
Collapse
Affiliation(s)
- Heng Jiang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Junjie Yang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yanfei Du
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
5
|
Gandhi H, Mahant S, Sharma AK, Kumar D, Dua K, Chellappan DK, Singh SK, Gupta G, Aljabali AAA, Tambuwala MM, Kapoor DN. Exploring the therapeutic potential of naturally occurring piceatannol in non-communicable diseases. Biofactors 2024; 50:232-249. [PMID: 37702264 DOI: 10.1002/biof.2009] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 08/28/2023] [Indexed: 09/14/2023]
Abstract
Piceatannol is a naturally occurring hydroxylated resveratrol analogue that can be found in a variety of fruits and vegetables. It has been documented to have a wide range of beneficial effects, including anti-inflammatory, antioxidant, anti-aging, anti-allergic, antidiabetic, neuroprotective, cardioprotective, and chemopreventive properties. Piceatannol has significantly higher antioxidant activity than resveratrol. Piceatannol has been shown in preclinical studies to have the ability to inhibit or reduce the growth of cancers in various organs such as the brain, breast, lung, colon, cervical, liver, prostate, and skin. However, the bioavailability of Piceatannol is comparatively lower than resveratrol and other stilbenes. Several approaches have been reported in recent years to enhance its bioavailability and biological activity, and clinical trials are required to validate these findings. This review focuses on several aspects of natural stilbene Piceatannol, its chemistry, and its mechanism of action, and its promising therapeutic potential for the prevention and treatment of a wide variety of complex human diseases.
Collapse
Affiliation(s)
- Himanshu Gandhi
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Shikha Mahant
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Abhishek Kumar Sharma
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Deepak Kumar
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
| | | | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
- Center for Transdisciplinary Research, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, England, UK
| | - Deepak N Kapoor
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, India
| |
Collapse
|
6
|
Fang J, Guan H. γ-Secretase inhibitor alleviates lipopolysaccharide-induced myocardial injury through regulating JAK2/STAT3 signaling. ENVIRONMENTAL TOXICOLOGY 2024; 39:135-147. [PMID: 37671635 DOI: 10.1002/tox.23962] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 07/17/2023] [Accepted: 08/22/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND Septic myocardial injury is one of the most life-threatening organ dysfunction. The γ-secretase-based approaches have been developed as potential strategies for diverse diseases management. Unfortunately, the role of γ-secretase inhibitor in septic myocardial injury is unclarified. The present study aims to investigate the effect of γ-secretase inhibitor in septic myocardial injury and reveal its mechanism. METHODS The mouse model of septic myocardial injury was established by intraperitoneally injection of lipopolysaccharide (LPS), and γ-secretase inhibitor MW167 was applied in this model. RNA-sequencing analysis and further bioinformatics analyses were used to screen differential expressed genes (DEGs) and potentially enriched pathways between LPS and LPS + MW167 mice. Pathological examination was performed using haematoxylin and eosin (HE) staining. SD-1029 was used to block JAK2/STAT3 signaling in H9C2 cells and western blot analysis quantified JAK2/STAT3-related proteins. RESULTS LPS induced myocardial injury accompanied with significant inflammatory infiltration and more apoptotic cells. Transcriptome sequencing screened 36 DEGs and bioinformatics identified JAK2/STAT3 signaling pathway was significantly enriched. Further in vitro experiments showed that γ-secretase inhibitor MW167 activated JAK2/STAT3 pathway. Additionally, MW167 restored cell viability, decreased myocardial injury markers including cardiac troponin I (cTnI) and brain natriuretic peptide (BNP), inhibited pro-inflammatory cytokines such as interleukin (IL)-1β and tumor necrosis factor-α (TNF-α) and reduced nitric oxide (NO), cyclooxygenase-2 (COX2) and inducible nitric oxide synthase (iNOS) release. Application of SD-1029 reversely deteriorated LPS-induced myocardial injury and inflammatory response in γ-secretase inhibitor-treated myocardial cells. CONCLUSION The results demonstrate that γ-secretase inhibitor alleviates septic myocardial injury via activating JAK2/STAT3 signaling, and provide novel therapeutic direction for septic myocardial injury.
Collapse
Affiliation(s)
- Jingyun Fang
- Department of Emergency, Ganzhou People's Hospital, Ganzhou, China
| | - Huan Guan
- Department of Emergency, Ganzhou People's Hospital, Ganzhou, China
| |
Collapse
|
7
|
Xie L, Zhou C, Wu Y, Fu X, Zhang G, Han X, Xie S, Chen G, Xu H, Deng B, Liu B, Zhou Y, Li A. Wenqingyin suppresses ferroptosis in the pathogenesis of sepsis-induced liver injury by activating the Nrf2-mediated signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154748. [PMID: 36933519 DOI: 10.1016/j.phymed.2023.154748] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/08/2023] [Accepted: 03/04/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Wenqingyin (WQY) is a classic traditional Chinese medicine formula used to treat various inflammatory diseases. However, its protective activity against ferroptosis in the pathogenesis of sepsis-induced liver injury and underlying mechanisms remain unclear. PURPOSE This study aimed to determine the therapeutic efficacy and potential mechanism of action of WQY in sepsis-induced liver injury both in vivo and in vitro. METHODS In vivo: Lipopolysaccharide was intraperitoneally injected into nuclear factor erythroid 2-related factor 2 (Nrf2) knockout (Nrf2-/-) and wild-type mice to construct a septic liver injury mouse model. Experimental mice were intraperitoneally injected with ferroptosis-1 and intragastrically administered WQY. In vitro: LO2 hepatocytes were stimulated with erastin to activate ferroptosis and later treated with varying concentrations of WQY and an Nrf2 inhibitor (ML385). Pathological damage was evaluated following hematoxylin and eosin staining. Lipid peroxidation levels were assessed using malondialdehyde, superoxide dismutase, and glutathione, as well as reactive oxygen species fluorescent probes. JC-1 staining was performed to evaluate the mitochondrial membrane potential damage. Quantitative reverse transcription polymerase chain reaction and western blot assay were performed to detect the related gene and protein levels. The levels of inflammatory factors were measured using Enzyme-Linked Immunosorbent Assay kits. RESULTS In vivo, sepsis-induced liver injury activated ferroptosis in mouse liver tissue. Fer-1 and WQY attenuated septic liver injury, which was associated with increased Nrf2 expression. Deletion of the Nrf2 gene led to aggravation of septic liver injury. The effect of WQY on the attenuation of septic liver injury was partially abolished by the knockdown of Nrf2. In vitro, erastin-induced ferroptosis resulted in decreased hepatocyte viability, lipid peroxidation, and mitochondrial membrane potential damage. WQY protected hepatocytes from erastin-induced ferroptosis by activating Nrf2. The attenuation effect of ferroptosis in hepatocytes by WQY was partially abolished by the inhibition of Nrf2. CONCLUSION Ferroptosis has a critical role in the development of sepsis-mediated liver injury. Inhibition of ferroptosis is a possible novel treatment strategy for alleviating septic liver injury. WQY attenuates sepsis-mediated liver injury by suppressing ferroptosis in hepatocytes, which is related to its ability to activate Nrf2.
Collapse
Affiliation(s)
- Lingpeng Xie
- Department of Hepatology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China
| | - Chuying Zhou
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuting Wu
- Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Xiuqiong Fu
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, 999077, China
| | - Guoyong Zhang
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xin Han
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shuwen Xie
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Guanghong Chen
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Honglin Xu
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bo Deng
- Department of Traditional Chinese Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510260, China.
| | - Yingchun Zhou
- School of Traditional Chinese Medicine, Department of Traditional Chinese Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Aimin Li
- Department of Hepatology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510315, China.
| |
Collapse
|
8
|
Chu W, Li YL, Li JJ, Lin J, Li M, Wang J, He JZ, Zhang YM, Yao J, Jin XJ, Cai H, Liu YQ. Guiqi Baizhu prescription ameliorates cytarabine-induced intestinal mucositis by targeting JAK2 to inhibit M1 macrophage polarization. Biomed Pharmacother 2023; 164:114902. [PMID: 37209628 DOI: 10.1016/j.biopha.2023.114902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Intestinal mucositis (IM) is characterized by damage to the intestinal mucosa resulting from inhibition of epithelial cell division and loss of renewal capacity following anticancer chemotherapy and radiotherapy. Cytarabine (Ara-C), the main chemotherapy drug for the treatment of leukemia and lymphoma, is a frequent cause of IM. Guiqi Baizhu prescription (GQBZP) is a traditional Chinese medicine with anti-cancer and anti-inflammatory effects. PURPOSE To determine if GQBZP can ameliorate Ara-C induced IM and identify and characterize the pharmacologic and pharmacodynamic mechanisms. STUDY DESIGN AND METHODS IM was induced in mice with Ara-C and concurrently treated with orally administered GQBZP. Body weight and food intake was monitored, with HE staining to calculate ileal histomorphometric scoring and villus length/crypt depth. Immunoblotting was used to detect intestinal tissue inflammatory factors. M1 macrophages (M1) were labeled with CD86 by flow cytometry and iNOS + F4/80 by immunofluorescence. Virtual screening was used to find potentially active compounds in GQBZP that targeted JAK2. In vitro, RAW264.7 cells were skewed to M1 macrophage polarization by lipopolysaccharide (LPS) and interferon-γ (INF-γ) and treated orally with GQBZP or potential active compounds. M1 was labeled with CD86 by flow cytometry and iNOS by immunofluorescence. ELISA was used to detect inflammatory factor expression. Active compounds against JAK2, p-JAK2, STAT1 and p-STAT1 were identified by western blotting and HCS fluorescence. Molecular dynamics simulations and pharmacokinetic predictions were carried out on representative active compounds. RESULTS Experimental results with mice in vivo suggest that GQBZP significantly attenuated Ara-C-induced ileal damage and release of pro-inflammatory factors by inhibiting macrophage polarization to M1. Molecular docking was used to identify potentially active compounds in GQBZP that targeted JAK2, a key factor in macrophage polarization to M1. By examining the main components of each herb and applying Lipinski's rules, ten potentially active compounds were identified. In vitro experimental results suggested that all 10 compounds of GQBZP targeted JAK2 and could inhibit M1 polarization in RAW264.7 cells treated with LPS and INF-γ. Among them, acridine and senkyunolide A down-regulated the expression of JAK2 and STAT1. MD simulations revealed that acridine and senkyunolide A were stable in the active site of JAK2 and exhibited good interactions with the surrounding amino acids. CONCLUSIONS GQBZP can ameliorate Ara-C-induced IM by reducing macrophage polarization to M1, and acridine and senkyunolide A are representative active compounds in GQBZP that target JAK2 to inhibit M1 polarization. Targeting JAK2 to regulate M1 polarization may be a valuable therapeutic strategy for IM.
Collapse
Affiliation(s)
- Wei Chu
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Ya-Ling Li
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China; Key Laboratory of Dun Huang Medical and Transformation, Ministry of Education of The People's Republic of China, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Jun-Jie Li
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Jia Lin
- College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Mi Li
- College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Jiao Wang
- College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Jian-Zheng He
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Yue-Mei Zhang
- Ophthalmology Department, First Hospital of Lanzhou University, 730000 Lanzhou, China
| | - Juan Yao
- College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China
| | - Xiao-Jie Jin
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China; College of Pharmacy, Gansu University of Chinese Medicine, 730000 Lanzhou, China.
| | - Hui Cai
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, 730000 Lanzhou, China; NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, 730000 Lanzhou, China.
| | - Yong-Qi Liu
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, 730000 Lanzhou, China; Key Laboratory of Dun Huang Medical and Transformation, Ministry of Education of The People's Republic of China, Gansu University of Chinese Medicine, 730000 Lanzhou, China.
| |
Collapse
|
9
|
Areny-Balagueró A, Solé-Porta A, Camprubí-Rimblas M, Campaña-Duel E, Ceccato A, Roig A, Closa D, Artigas A. Bioengineered extracellular vesicles: future of precision medicine for sepsis. Intensive Care Med Exp 2023; 11:11. [PMID: 36894763 PMCID: PMC9998145 DOI: 10.1186/s40635-023-00491-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/01/2023] [Indexed: 03/11/2023] Open
Abstract
Sepsis is a syndromic response to infection and is frequently a final common pathway to death from many infectious diseases worldwide. The complexity and high heterogeneity of sepsis hinder the possibility to treat all patients with the same protocol, requiring personalized management. The versatility of extracellular vesicles (EVs) and their contribution to sepsis progression bring along promises for one-to-one tailoring sepsis treatment and diagnosis. In this article, we critically review the endogenous role of EVs in sepsis progression and how current advancements have improved EVs-based therapies toward their translational future clinical application, with innovative strategies to enhance EVs effect. More complex approaches, including hybrid and fully synthetic nanocarriers that mimic EVs, are also discussed. Several pre-clinical and clinical studies are examined through the review to offer a general outlook of the current and future perspectives of EV-based sepsis diagnosis and treatment.
Collapse
Affiliation(s)
- Aina Areny-Balagueró
- Institut d’Investigació i Innovació Parc Taulí (I3PT), Parc Taulí Hospital Universitari, 08208 Sabadell, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Anna Solé-Porta
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - Marta Camprubí-Rimblas
- Institut d’Investigació i Innovació Parc Taulí (I3PT), Parc Taulí Hospital Universitari, 08208 Sabadell, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Present Address: Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias, CIBERES-Instituto De Salud Carlos III, 28029 Madrid, Spain
| | - Elena Campaña-Duel
- Institut d’Investigació i Innovació Parc Taulí (I3PT), Parc Taulí Hospital Universitari, 08208 Sabadell, Spain
| | - Adrián Ceccato
- Institut d’Investigació i Innovació Parc Taulí (I3PT), Parc Taulí Hospital Universitari, 08208 Sabadell, Spain
- Present Address: Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias, CIBERES-Instituto De Salud Carlos III, 28029 Madrid, Spain
| | - Anna Roig
- Institut de Ciència de Materials de Barcelona, ICMAB-CSIC, Campus UAB, 08193 Bellaterra, Spain
| | - Daniel Closa
- Institut d’Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| | - Antonio Artigas
- Institut d’Investigació i Innovació Parc Taulí (I3PT), Parc Taulí Hospital Universitari, 08208 Sabadell, Spain
- Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Present Address: Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias, CIBERES-Instituto De Salud Carlos III, 28029 Madrid, Spain
- Servei de Medicina Intensiva, Corporació Sanitària i Universitària Parc Taulí, 08208 Sabadell, Spain
| |
Collapse
|
10
|
Li J, Huang P, Cheng W, Niu Q. Stilbene-based derivatives as potential inhibitors of trimethylamine (TMA)-lyase affect gut microbiota in coronary heart disease. Food Sci Nutr 2023; 11:93-100. [PMID: 36655110 PMCID: PMC9834892 DOI: 10.1002/fsn3.3046] [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: 12/06/2021] [Revised: 08/05/2022] [Accepted: 08/14/2022] [Indexed: 01/21/2023] Open
Abstract
Coronary heart disease (CHD) is defined by atherosclerosis, which may result in stenosis or blockage of the arterial cavity, leading to ischemic cardiac diseases such as angina and myocardial infarction (MI). Accumulating evidence indicates that the gut microbiota play a critical role in the initiation and progression of CHD. The gut microbial metabolite trimethylamine N-oxide (TMAO) is intimately linked to the pathophysiology of CHD. The hepatic flavin-containing monooxygenases (FMOs) convert trimethylamine (TMA) to TMAO. As a result, it is critical to prevent TMA generation. Stilbenes could reduce cardiovascular disease mortality. Twelve stilbenes with inhibitory activity against TMA-lyase were compiled and evaluated in this study. Docking results showed Resveratroloside had the highest Vina score, indicating that it was the most active and might be employed as a lead molecule for further structural modification.
Collapse
Affiliation(s)
- Jincai Li
- School of Traditional Chinese MedicineBozhou UniversityBozhouChina
| | - Peng Huang
- School of PharmacyAnhui University of Chinese MedicineHefeiChina
| | - Wangxing Cheng
- School of PharmacyAnhui University of Chinese MedicineHefeiChina
| | - Qian Niu
- Department of PharmacyBozhou Vocational and Technical CollegeBozhouChina
| |
Collapse
|
11
|
Omaveloxolone attenuates the sepsis-induced cardiomyopathy via activating the nuclear factor erythroid 2-related factor 2. Int Immunopharmacol 2022; 111:109067. [PMID: 35908503 DOI: 10.1016/j.intimp.2022.109067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 12/14/2022]
Abstract
Sepsis-induced cardiomyopathy (SIC) is a common complication of sepsis and is the main reason for the high mortality in sepsis patients. More recent studies have indicated that activating nuclear factor erythroid 2-related factor 2 (Nrf2) signaling plays a protective role in SIC. As a potent activator of Nrf2, Omaveloxolone plays a pivotal role in defending against oxidative stress and the inflammatory response. Thus, we examined the efficacy of omaveloxolone in SIC. In the present study, the mice were injected intraperitoneally with a single dose of LPS (10 mg/kg) for 12 h to induce SIC. The data in our study indicated that omaveloxolone administration significantly improved cardiac injury and dysfunction in LPS-induced SIC. In addition, omaveloxolone administration reduced SIC-related cardiac oxidative stress, the inflammatory response and cardiomyocyte apoptosis in mice. In addition, omaveloxolone administration also improved LPS-induced cardiomyocyte injury in an in vitro model using H9C2 cells. Moreover, knockdown of Nrf2 by si-Nrf2 abolished the omaveloxolone-mediated cardioprotective effects. In conclusion, omaveloxolone has potent cardioprotective potential in treating sepsis and SIC via activation of the Nrf2 signaling pathway.
Collapse
|
12
|
Piceatannol-mediated JAK2/STAT3 signaling pathway inhibition contributes to the alleviation of oxidative injury and collagen synthesis during pulmonary fibrosis. Int Immunopharmacol 2022; 111:109107. [PMID: 35932616 DOI: 10.1016/j.intimp.2022.109107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 11/21/2022]
Abstract
Pulmonary fibrosis (PF) is characterized by oxidative injury and excessive collagen synthesis in lung fibroblasts, causing impaired pulmonary function and chronic lung injury. Piceatannol, a dietary polyphenol, possesses vital pharmacological effects in metabolic disorders, cancers, cardiovascular disease and infectious disease; however, its role in PF is still not completely elucidated. Mice (8 to 10 weeks old) were administered bleomycin (BLM) intratracheally (2 U/kg) to establish an in vivo PF model. Murine primary lung fibroblasts were isolated and stimulated with TGF-β (10 ng/mL) for 48 h to induce its activation. Meanwhile, mice or primary lung fibroblasts were treated with different doses of piceatannol to observe its protective roles. Pulmonary function and arterial blood gas were detected to assess pulmonary physiological status. Collagen deposition and the mRNA levels of profibrotic genes were determined by H&E staining and RT-PCR. Meanwhile, the protein and mRNA markers, as well as end-product of oxidative stress were detected in vivo and in vitro. The results showed that pulmonary function was significantly impaired in BLM-induced mice, accompanied by elevated oxidative stress and excessive collagen synthesis. Piceatannol significantly improved pulmonary function and decreased oxidative injury as well as collagen synthesis in mice with PF. Mechanically, piceatannol treatment significantly inhibited the activation of JAK2/STAT3 signaling pathway in BLM-induced mice and TGF-β-induced lung fibroblasts. Additional findings also demonstrated that coumermycin A1 (C-A1), an agonist of JAK2, could abolish the effects of piceatannol on TGF-β-induced lung fibroblasts and reactivated the phosphorylation STAT3. Taken together, our study demonstrated that piceatannol could protect against oxidative injury and collagen synthesis during PF in a JAK2/STAT3 signaling pathway-dependent manner.
Collapse
|
13
|
Gao L, Zhai Z, Shi Q, Yan J, Zhou L, Wu Y, Zeng Q, Tian G, Li H. MiR-501-5p alleviates cardiac dysfunction in septic patients through targeting NR4A3 to prevent its binding with Bcl-2. Cell Cycle 2022; 21:961-971. [PMID: 35230891 PMCID: PMC9037443 DOI: 10.1080/15384101.2022.2035618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Sepsis-induced myocardial dysfunction is a common complication in septic patients. To date, a limited number of biomarkers that could predict cardiomyocyte apoptosis have been explored. In this study, we successfully established a cecal ligation and puncture (CLP)-induced septic model, and it was found that miR-501-5p expression was down-regulated in peripheral blood samples of septic patients with cardiac dysfunction, lipopolysaccharide (LPS)-induced cardiomyocytes, and the myocardium and peripheral blood in the septic model. Moreover, it was revealed that miR-501-5p overexpression could increase left ventricular diastolic pressure (LVDP), fractional shortening (FS), ejection fraction (EF), and maximum rate of the rise of left ventricular pressure (+dp/dt) in vivo, while it decreased the levels of myocardial injury-related indicators. In addition, LPS induction accelerated apoptosis and elevated the inflammation in HL-1 and HCM cells, which could be reversed by miR-501-5p overexpression. Mechanistically, we considered nuclear receptor subfamily 4 group A member 3 (NR4A3) as the target of miR-501-5p, and it was found that miR-501-5p prevented the binding between NR4A3 and Bcl-2. It was found that miR-501-5p exerted an inhibitory effect on cardiomyocyte apoptosis and inflammation in a NR4A3-dependent manner. Overall, our results may provide evidence for consideration of miR-501-5p in the therapy of sepsis.
Collapse
Affiliation(s)
- Lan Gao
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Zhongjie Zhai
- Department of Health Statistics, Faculty of Preventive Medicine, Air Force Medical University, Xi’an, Shaanxi, China
| | - Qindong Shi
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jinqi Yan
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Linjing Zhou
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yongxin Wu
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Qinjing Zeng
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Gang Tian
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China,CONTACT Gang Tian Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Hao Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China,Hao Li Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, Xi'an, China
| |
Collapse
|