1
|
Chen F, Chen R, Yang L, Shen B, Wang Y, Gao Y, Tan R, Zhao X. Magnesium-assisted hydrogen improves isoproterenol-induced heart failure. Med Gas Res 2025; 15:459-470. [PMID: 40300881 DOI: 10.4103/mgr.medgasres-d-24-00135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/19/2025] [Indexed: 05/01/2025] Open
Abstract
Heart failure (HF) is a leading cause of mortality among patients with cardiovascular disease and is often associated with myocardial apoptosis and endoplasmic reticulum stress (ERS). While hydrogen has demonstrated potential in reducing oxidative stress and ERS, recent evidence suggests that magnesium may aid in hydrogen release within the body, further enhancing these protective effects. This study aimed to investigate the cardioprotective effects of magnesium in reducing apoptosis and ERS through hydrogen release in a rat model of isoproterenol (ISO)-induced HF. Magnesium was administered orally to ISO-induced HF rats, which improved cardiac function, reduced myocardial fibrosis and cardiac hypertrophy, and lowered the plasma levels of creatine kinase-MB, cardiac troponin-I, and N-terminal B-type natriuretic peptide precursor in ISO-induced HF rats. It also inhibited cardiomyocyte apoptosis by upregulating B-cell lymphoma-2, downregulating Bcl-2-associated X protein, and suppressing ERS markers (glucose-related protein 78, activating transcription factor 4, and C/EBP-homologous protein). Magnesium also elevated hydrogen levels in blood, plasma, and cardiac tissue, as well as in artificial gastric juice and pure water, where hydrogen release lasted for at least four hours. Additionally, complementary in vitro experiments were conducted using H9C2 cardiomyocyte injury models, with hydrogen-rich culture medium as the intervention. Hydrogen-rich culture medium improved the survival and proliferation of ISO-treated H9C2 cells, reduced the cell surface area, inhibited apoptosis, and downregulated ERS pathway proteins. However, the protective effects of hydrogen were negated by tunicamycin (an inducer of ERS) in H9C2 cells. In conclusion, magnesium exerts significant cardioprotection by mitigating ERS and apoptosis through hydrogen release effects in ISO-induced HF.
Collapse
Affiliation(s)
- Fengbao Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Ruimin Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Lili Yang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
- New Drug Evaluation Center of Shandong Academy of Pharmaceutical Sciences, Shandong Academy of Pharmaceutical Sciences, Ji'nan, Shandong Province, China
| | - Bowen Shen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
- School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Yunting Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Yongfeng Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Rui Tan
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Xiaomin Zhao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| |
Collapse
|
2
|
Jin J, Yue L, Du M, Geng F, Gao X, Zhou Y, Lu Q, Pan X. Molecular Hydrogen Therapy: Mechanisms, Delivery Methods, Preventive, and Therapeutic Application. MedComm (Beijing) 2025; 6:e70194. [PMID: 40297245 PMCID: PMC12035766 DOI: 10.1002/mco2.70194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Molecular hydrogen (H2), recognized as the smallest gas molecule, is capable of permeating cellular membranes and diffusing throughout the body. Due to its high bioavailability, H2 is considered a therapeutic gas for the treatment of various diseases. The therapeutic efficacy of hydrogen is contingent upon factors such as the administration method, duration of contact with diseased tissue, and concentration at targeted sites. H2 can be administered exogenously and is also produced endogenously within the intestinal tract. A comprehensive understanding of its delivery mechanisms and modes of action is crucial for advancing hydrogen medicine. This review highlights H₂'s mechanisms of action, summarizes its administration methods, and explores advancements in treating intestinal diseases (e.g., inflammatory bowel disease, intestinal ischemia-reperfusion, colorectal cancer). Additionally, its applications in managing other diseases are discussed. Finally, the challenges associated with its clinical application and potential solutions are explored. We propose that current delivery challenges faced by H2 can be effectively addressed through the use of nanoplatforms; furthermore, interactions between hydrogen and gut microbiota may provide insights into its mechanisms for treating intestinal diseases. Future research should explore the synergistic effects of H2 in conjunction with conventional therapies and develop personalized treatment plans to achieve precision medicine.
Collapse
Affiliation(s)
- Jiayi Jin
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Lijun Yue
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Maoru Du
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Feng Geng
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Xue Gao
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Yuming Zhou
- Department of Laboratory MedicineYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Qianqian Lu
- Department of OncologyYantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Xiaohong Pan
- School of PharmacyBinzhou Medical UniversityYantaiChina
| |
Collapse
|
3
|
Denver P, Cunningham C. Microglial activation and neuroinflammation in acute and chronic cognitive deficits in sepsis. Neuropharmacology 2025; 267:110285. [PMID: 39746541 DOI: 10.1016/j.neuropharm.2024.110285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/11/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Sepsis is characterised by dysregulated immune responses to infection, leading to multi-organ dysfunction and high rates of mortality. With increasing survival rates in recent years long-term neurological and psychiatric consequences have become more apparent in survivors. Many patients develop sepsis associated encephalopathy (SAE) which encompasses the profound but usually transient neuropsychiatric syndrome delirium but also new brain injury that emerges in the months and years post-sepsis. It is now clear that systemic inflammatory signals reach the brain during sepsis and that very significant neuroinflammation ensues. The major brain resident immune cell population, the microglia, has been implicated in acute and chronic cognitive dysfunction in animal models of sepsis based on a growing number of studies using bacterial endotoxin and in polymicrobial sepsis models such as cecal ligation and puncture. The current review explores the effects of sepsis on the brain, focussing on how systemic insults translate to microglial activation and neuroinflammation and how this disrupts neuronal function and integrity. We examine what has been demonstrated specifically with respect to microglial activation, revealing robust evidence for a role for neuroinflammation in sepsis-induced brain sequelae but less clear information on the extent of the specific microglial contribution to this, arising from findings using global knockout mice, non-selective drugs and treatments that equally target peripheral and central compartments. There is, nonetheless, clear evidence that microglia do become activated and do contribute to brain consequences of sepsis thus arguing for improved understanding of these neuroinflammatory processes toward the prevention and treatment of sepsis-induced brain dysfunction.
Collapse
Affiliation(s)
- Paul Denver
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, Dublin 2, Ireland.
| |
Collapse
|
4
|
Irzan FI, Retinasamy T, Wen WR, Sheng ETM, Shaikh MF, Arulsamy A. The Role of HMGB1 in Infection-Related Cognitive Deficits. FRONT BIOSCI-LANDMRK 2025; 30:25544. [PMID: 40018921 DOI: 10.31083/fbl25544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 03/01/2025]
Abstract
Infectious diseases caused by fungi, viruses, or bacteria can have a profound impact on human cognition. This can be due to either direct spread to the central nervous system (CNS) or indirect neuroinflammation. Ultimately causing neuronal damage and even neurodegeneration. Deteriorations in cognition, such as poor encoding and attention deficits, have been reported secondary to infectious diseases. Preclinical studies have identified the underlying mechanisms of these infection-related cognitive effects, such as through blood-brain barrier (BBB) disruption and M1 microglial polarization. These mechanisms are spearheaded by inflammatory markers that are released/initiated by the pathogens over the course of the infection. Among them, the high mobility group box 1 (HMGB1) protein is a common biomarker implicated across several infection-related cognitive deficits. Understanding these effects and mechanisms is crucial for the development of strategies to prevent and treat infection-related cognitive impairment. This review will thus consolidate and elucidate the current knowledge on the potential role of HMGB1 as a therapeutic target for infection-related cognitive impairments. This review will not only advance scientific understanding but also have significant clinical and public health implications, especially considering recent global health challenges. Based on the selected articles, extracellular HMGB1, as opposed to intracellular HMGB1, acts as damage-associated molecular patterns (DAMPs) or alarmins when released in the peripheries secondary to inflammasome activation. Due to their low molecular weight, they then enter the CNS through routes such as retrograde transport along the afferent nerves, or simple diffusion across the impaired BBB. This results in further disruption of the brain microenvironment due to the dysregulation of other regulatory pathways. The outcome is structural neuronal changes and cognitive impairment. Given its key role in neuroinflammation, HMGB1 holds promise as both a biomarker for diagnostic detection and a potential therapeutic target candidate for preventing infection-related cognitive impairment.
Collapse
Affiliation(s)
- Fathima Ijaza Irzan
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Thaarvena Retinasamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Wong Ruo Wen
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Edward Ting Ming Sheng
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
- School of Dentistry and Medical Sciences, Charles Sturt University, Orange, NSW 2800, Australia
| | - Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Petaling Jaya, Selangor, Malaysia
| |
Collapse
|
5
|
Nakayama M, Watanabe K, Sato E, Ito Y, Kadota N, Konishi K, Aizawa C, Maruyama Y, Fujimaru T, Nagahama M, Taki F, Suzuki M. Hemodialysis employing molecular hydrogen (H 2) enriched dialysis solution may improve dialysis related fatigue through impact on energy metabolism. Sci Rep 2025; 15:5039. [PMID: 39934143 PMCID: PMC11814270 DOI: 10.1038/s41598-025-88827-2] [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: 08/16/2024] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Hemodialysis employing molecular hydrogen (H2)-enriched dialysis solution rendered by water electrolysis (E-HD), has been reported to alleviate dialysis-related fatigue, but its association with metabolic profiles remains unclear. Eighty-one patients undergoing standard HD were classified into 3 groups [Group A (n = 25, 30.9%): fatigue with activity reduction-subgroups A1: chronic persistent fatigue (n = 11), A2: fatigue only on dialysis days (n = 14); Group B: fatigue without activity reduction (n = 24, 29.6%); Group C (n = 32, 39.5%): no fatigue], and their changes in fatigue, body composition, and metabolic profiles were studied following 12 months of E-HD. There were no significant differences in baseline characteristics among the groups. Over the 12 months after E-HD initiation, fatigue in Group A significantly decreased, while no changes in Group-B and C. Bio-impedance analysis revealed no significant changes in A1, but significant reductions in body fat and increases in skeletal muscle mass were observed despite no significant weight change in A2. Enrichment analysis suggested significant differences in metabolic pathways such as fatty acid metabolism, citric acid cycle, and glycolysis between Groups A and C at baseline, and these differences were mitigated by E-HD. E-HD could suppress dialysis-related fatigue, through possible involvement of altered energy metabolism of patients. E-HD may represent a new paradigm for uremia treatment beyond traditional solute removal-based dialysis therapies.
Collapse
Affiliation(s)
- Masaaki Nakayama
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan.
- Department of Research Management, St Luke's International University, Tokyo, Japan.
- Head Office for Open Innovation Business Development Strategy, Tohoku University, Sendai, Japan.
| | - Kimio Watanabe
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Emiko Sato
- Division of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yugo Ito
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Nozomi Kadota
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Kasumi Konishi
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Chiharu Aizawa
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Yukio Maruyama
- Department of Kidney and Hypertension, The Tokyo Jikei University School of Medicine, Tokyo, Japan
| | - Takuya Fujimaru
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Masahiko Nagahama
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Fumika Taki
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| | - Michiko Suzuki
- Kidney Center, St Luke's International Hospital, 9-1 Akashi-cho, Chuo-ku, Tokyo, 104-8560, Japan
| |
Collapse
|
6
|
Yang L, Li J, Liu F, Chai X, Fang Z, Zhang X. The Biological Changes of Synaptic Plasticity in the Pathological Process of Sepsis-associated Encephalopathy. Curr Neuropharmacol 2025; 23:359-374. [PMID: 39473252 DOI: 10.2174/1570159x23666241028105746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/24/2024] [Accepted: 02/14/2024] [Indexed: 03/25/2025] Open
Abstract
Sepsis-associated encephalopathy (SAE) is a form of cognitive and psychological impairment resulting from sepsis, which occurs without any central nervous system infection or structural brain injury. Patients may experience long-term cognitive deficits and psychiatric disorders even after discharge. However, the underlying mechanism remains unclear. As cognitive function and mental disease are closely related to synaptic plasticity, it is presumed that alterations in synaptic plasticity play an essential role in the pathological process of SAE. Here, we present a systematic description of the pathogenesis of SAE, which is primarily driven by glial cell activation and subsequent release of inflammatory mediators. Additionally, we elucidate the alterations in synaptic plasticity that occur during SAE and comprehensively discuss the roles played by glial cells and inflammatory factors in this process. In this review, we mainly discuss the synaptic plasticity of SAE, and the main aim is to show the consequences of SAE on inflammatory factors and how they affect synaptic plasticity. This review may enhance our understanding of the mechanism underlying cognitive dysfunction and provide valuable insights into identifying appropriate therapeutic targets for SAE.
Collapse
Affiliation(s)
- Lin Yang
- Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Jin Li
- Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Department of Critical Care Medicine, Air Force Medical Center, Beijing, 100142, China
| | - Fuhong Liu
- Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Xin Chai
- Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Zongping Fang
- Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- The Third Department of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xijing Zhang
- Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| |
Collapse
|
7
|
Cazalla E, Cuadrado A, García-Yagüe ÁJ. Role of the transcription factor NRF2 in maintaining the integrity of the Blood-Brain Barrier. Fluids Barriers CNS 2024; 21:93. [PMID: 39574123 PMCID: PMC11580557 DOI: 10.1186/s12987-024-00599-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 11/15/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND The Blood-Brain Barrier (BBB) is a complex and dynamic interface that regulates the exchange of molecules and cells between the blood and the central nervous system. It undergoes structural and functional throughout oxidative stress and inflammation, which may compromise its integrity and contribute to the pathogenesis of neurodegenerative diseases. MAIN BODY Maintaining BBB integrity is of utmost importance in preventing a wide range of neurological disorders. NRF2 is the main transcription factor that regulates cellular redox balance and inflammation-related gene expression. It has also demonstrated a potential role in regulating tight junction integrity and contributing to the inhibition of ECM remodeling, by reducing the expression of several metalloprotease family members involved in maintaining BBB function. Overall, we review current insights on the role of NRF2 in addressing protection against the effects of BBB dysfunction, discuss its involvement in BBB maintenance in different neuropathological diseases, as well as, some of its potential activators that have been used in vitro and in vivo animal models for preventing barrier dysfunction. CONCLUSIONS Thus, emerging evidence suggests that upregulation of NRF2 and its target genes could suppress oxidative stress, and neuroinflammation, restore BBB integrity, and increase its protection.
Collapse
Affiliation(s)
- Eduardo Cazalla
- Department of Biochemistry, School of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Sols-Morreale" (CSIC-UAM), C/ Arturo Duperier, 4, Madrid, 28029, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Antonio Cuadrado
- Department of Biochemistry, School of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain
- Instituto de Investigaciones Biomédicas "Sols-Morreale" (CSIC-UAM), C/ Arturo Duperier, 4, Madrid, 28029, Spain
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ángel Juan García-Yagüe
- Department of Biochemistry, School of Medicine, Autonomous University of Madrid (UAM), Madrid, Spain.
- Instituto de Investigaciones Biomédicas "Sols-Morreale" (CSIC-UAM), C/ Arturo Duperier, 4, Madrid, 28029, Spain.
- Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
8
|
Mohamed AR, Fares NH, Mahmoud YI. Morin Ameliorates Lipopolysaccharides-Induced Sepsis-Associated Encephalopathy and Cognitive Impairment in Albino Mice. Neurochem Res 2024; 50:14. [PMID: 39549093 PMCID: PMC11568986 DOI: 10.1007/s11064-024-04269-8] [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: 03/16/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 11/18/2024]
Abstract
Sepsis-associated encephalopathy is a common neurological complication of sepsis that is characterized by neuroinflammation, oxidative stress and apoptosis, which results in cognitive impairments in septic survivors. Despite numerous treatment options for this condition, none of them are definite. Therefore, this study aimed to investigate the impact of morin, a flavone known for its neuroprotective and anti-inflammatory effects, against lipopolysaccharides-induced sepsis-associated encephalopathy in albino mice for 7 days. Mice were divided into 4 groups: Negative control, morin, septic, and septic morin-treated mice. Sepsis was induced by a single injection of lipopolysaccharides (5 mg/kg, intraperitoneally), morin (50 mg/kg b. wt.) was given orally, starting from 5 h after sepsis induction, then daily for 4 other days. Morin ameliorated septic structural and functional alternations as manifested by improving the survival rate, the behavioral functions, in addition to preserving and protecting the brain tissue. This was accompanied with the augmentation of the total antioxidant capacity, as well as the suppression of tissue levels of the lipid peroxidation marker malondialdehyde, apoptosis (cleaved-caspase-3), glial fibrillary acidic protein, and the proinflammatory cytokine tumor necrosis factor. In conclusion, morin has a promising ameliorative effect to counteract the sepsis-associated encephalopathy via its anti-inflammatory and antioxidant effects and to prevent the associated cognitive impairments.
Collapse
Affiliation(s)
- Asmaa R Mohamed
- Zoology Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Nagui H Fares
- Zoology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Yomna I Mahmoud
- Zoology Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
9
|
Xiao K, Liu J, Sun Y, Chen S, Ma J, Cao M, Yang Y, Pan Z, Li P, Du Z. Anti-inflammatory and antioxidant activity of high concentrations of hydrogen in the lung diseases: a systematic review and meta-analysis. Front Immunol 2024; 15:1444958. [PMID: 39211045 PMCID: PMC11357939 DOI: 10.3389/fimmu.2024.1444958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
As a small molecule, hydrogen is colorless, odorless and lightest. Many studies conducted that hydrogen can protect almost every organ, including the brain, heart muscle, liver, small intestine, and lungs. To verify whether high concentrations of hydrogen (HCH) has anti-inflammatory and antioxidant activities on respiratory system, we product a systematic review and meta-analysis. We investigated MEDLINE-PubMed, Cochrane Library, ScienceDirect, Wiley and SpringerLink database and selected in vivo studies related to the anti-inflammatory or antioxidant effects of HCH in the lung diseases which were published until September 2023. We firstly identified 437 studies and only 12 met the inclusion criteria. They all conducted in rodents. The results showed that HCH had a positive effect on the reduction of tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, IL-4, IL-8, malondialdehyde (MDA), superoxide dismutase (SOD) and reactive oxygen species (ROS); but there is no effect on IL-6, we speculated that may contribute to the test results for different body fluids and at different points in time. This meta-analysis discovered the protective effects on inflammation and oxidative stress, but whether there exists more effects on reduction of inflammatory and oxidant mediators needs to be further elucidated.
Collapse
Affiliation(s)
- Kang Xiao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jianwei Liu
- Public Health Monitoring and Evaluation Institute of Shandong Provincial Center for Disease Control and Prevention, Ji’nan, Shandong, China
| | - Yuxin Sun
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Shangya Chen
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Jiazi Ma
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Mao Cao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Yong Yang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Zhifeng Pan
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Peng Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Zhongjun Du
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| |
Collapse
|
10
|
Hu Q, Li Y, Lin Z, Zhang H, Chen H, Chao C, Zhao C. The Molecular Biological Mechanism of Hydrogen Therapy and Its Application in Spinal Cord Injury. Drug Des Devel Ther 2024; 18:1399-1414. [PMID: 38707612 PMCID: PMC11068043 DOI: 10.2147/dddt.s463177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024] Open
Abstract
Hydrogen, which is a novel biomedical molecule, is currently the subject of extensive research involving animal experiments and in vitro cell experiments, and it is gradually being applied in clinical settings. Hydrogen has been proven to possess anti-inflammatory, selective antioxidant, and antiapoptotic effects, thus exhibiting considerable protective effects in various diseases. In recent years, several studies have provided preliminary evidence for the protective effects of hydrogen on spinal cord injury (SCI). This paper provides a comprehensive review of the potential molecular biology mechanisms of hydrogen therapy and its application in treating SCI, with an aim to better explore the medical value of hydrogen and provide new avenues for the adjuvant treatment of SCI.
Collapse
Affiliation(s)
- Quan Hu
- Department of Neurosurgery, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Yingxiao Li
- Department of Gynecology, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Zhaochen Lin
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Hao Zhang
- Department of Rehabilitation Medical Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Haoyue Chen
- Department of Rehabilitation Medical Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Cui Chao
- Hydrogen Medical Research Center, The Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| | - Chuanliang Zhao
- Department of Orthopedics, the Affiliated Taian City Central Hospital of Qingdao University, Tai’an City, Shandong, 271000, People’s Republic of China
| |
Collapse
|
11
|
Zhu Q, Wan L, Huang H, Liao Z. IL-1β, the first piece to the puzzle of sepsis-related cognitive impairment? Front Neurosci 2024; 18:1370406. [PMID: 38665289 PMCID: PMC11043581 DOI: 10.3389/fnins.2024.1370406] [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: 01/14/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is a leading cause of death resulting from an uncontrolled inflammatory response to an infectious agent. Multiple organ injuries, including brain injuries, are common in sepsis. The underlying mechanism of sepsis-associated encephalopathy (SAE), which is associated with neuroinflammation, is not yet fully understood. Recent studies suggest that the release of interleukin-1β (IL-1β) following activation of microglial cells plays a crucial role in the development of long-lasting neuroinflammation after the initial sepsis episode. This review provides a comprehensive analysis of the recent literature on the molecular signaling pathways involved in microglial cell activation and interleukin-1β release. It also explores the physiological and pathophysiological role of IL-1β in cognitive function, with a particular focus on its contribution to long-lasting neuroinflammation after sepsis. The findings from this review may assist healthcare providers in developing novel interventions against SAE.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Wan
- Department of Medical Genetics/Prenatal Diagnostic Center Nursing and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhimin Liao
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Tian M, Zhan Y, Cao J, Gao J, Sun J, Zhang L. Targeting blood-brain barrier for sepsis-associated encephalopathy: Regulation of immune cells and ncRNAs. Brain Res Bull 2024; 209:110922. [PMID: 38458135 DOI: 10.1016/j.brainresbull.2024.110922] [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: 10/21/2023] [Revised: 02/14/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
Sepsis causes significant morbidity and mortality worldwide, most surviving patients show acute or chronic mental disorders, which are known as sepsis-associated encephalopathy (SAE). SAE involves many pathological processes, including the blood-brain barrier (BBB) damage. The BBB is located at the interface between the central nervous system and the surrounding environment, which protects the central nervous system (CNS) from the invasion of exogenous molecules, harmful substances or microorganisms in the blood. Recently, a growing number of studies have indicated that the BBB destruction was involved in SAE and played an important role in SAE-induced brain injury. In the present review, we firstly reveal the pathological processes of SAE such as the neurotransmitter disorders, oxidative stress, immune dysfunction and BBB destruction. Moreover, we introduce the structure of BBB, and describe the immune cells including microglia and astrocytes that participate in the BBB destruction after SAE. Furthermore, in view of the current research on non-coding RNAs (ncRNAs), we explain the regulatory mechanism of ncRNAs including long noncoding RNAs (lncRNAs), microRNAs (miRNAs) and circular RNAs (circRNAs) on BBB in the processes of SAE. Finally, we propose some challenges and perspectives of regulating BBB functions in SAE. Hence, on the basis of these effects, both immune cells and ncRNAs may be developed as therapeutic targets to protect BBB for SAE patients.
Collapse
Affiliation(s)
- Mi Tian
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, China
| | - Yunliang Zhan
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jinyuan Cao
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, China
| | - Jinqi Gao
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, China
| | - Jie Sun
- Department of Anesthesiology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu Province, China.
| | - Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
13
|
Nakayama M, Kabayama S, Miyazaki M. Application of Electrolyzed Hydrogen Water for Management of Chronic Kidney Disease and Dialysis Treatment-Perspective View. Antioxidants (Basel) 2024; 13:90. [PMID: 38247514 PMCID: PMC10812465 DOI: 10.3390/antiox13010090] [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: 12/22/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
Chronic kidney disease (CKD), which is globally on the rise, has become an urgent challenge from the perspective of public health, given its risk factors such as end-stage renal failure, cardiovascular diseases, and infections. The pathophysiology of CKD, including dialysis patients, is deeply associated with enhanced oxidative stress in both the kidneys and the entire body. Therefore, the introduction of a safe and widely applicable antioxidant therapy is expected as a measure against CKD. Electrolyzed hydrogen water (EHW) generated through the electrolysis of water has been confirmed to possess chemical antioxidant capabilities. In Japan, devices producing this water have become popular for household drinking water. In CKD model experiments conducted to date, drinking EHW has been shown to suppress the progression of kidney damage related to hypertension. Furthermore, clinical studies have reported that systemic oxidative stress in patients undergoing dialysis treatment using EHW is suppressed, leading to a reduction in the incidence of cardiovascular complications. In the future, considering EHW as one of the comprehensive measures against CKD holds significant importance. The medical utility of EHW is believed to be substantial, and further investigation is warranted.
Collapse
Affiliation(s)
- Masaaki Nakayama
- Kidney Center, St. Luke’s International Hospital, Tokyo 104-8560, Japan
- Division of Blood Purification, Tohoku University Hospital, Sendai 980-8574, Japan; (S.K.); (M.M.)
| | - Shigeru Kabayama
- Division of Blood Purification, Tohoku University Hospital, Sendai 980-8574, Japan; (S.K.); (M.M.)
- Graduate School of Science, Technology & Innovation, Kobe University, Kobe 657-8501, Japan
- Nihon Trim Co., Ltd., Osaka 530-0001, Japan
| | - Mariko Miyazaki
- Division of Blood Purification, Tohoku University Hospital, Sendai 980-8574, Japan; (S.K.); (M.M.)
- Division of Nephrology, Rheumatology and Endocrinology, Graduate School of Medicine, Tohoku University, Sendai 980-8574, Japan
| |
Collapse
|
14
|
Cheng D, Long J, Zhao L, Liu J. Hydrogen: A Rising Star in Gas Medicine as a Mitochondria-Targeting Nutrient via Activating Keap1-Nrf2 Antioxidant System. Antioxidants (Basel) 2023; 12:2062. [PMID: 38136182 PMCID: PMC10740752 DOI: 10.3390/antiox12122062] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
The gas molecules O2, NO, H2S, CO, and CH4, have been increasingly used for medical purposes. Other than these gas molecules, H2 is the smallest diatomic molecule in nature and has become a rising star in gas medicine in the past few decades. As a non-toxic and easily accessible gas, H2 has shown preventive and therapeutic effects on various diseases of the respiratory, cardiovascular, central nervous system, and other systems, but the mechanisms are still unclear and even controversial, especially the mechanism of H2 as a selective radical scavenger. Mitochondria are the main organelles regulating energy metabolism in living organisms as well as the main organelle of reactive oxygen species' generation and targeting. We propose that the protective role of H2 may be mainly dependent on its unique ability to penetrate every aspect of cells to regulate mitochondrial homeostasis by activating the Keap1-Nrf2 phase II antioxidant system rather than its direct free radical scavenging activity. In this review, we summarize the protective effects and focus on the mechanism of H2 as a mitochondria-targeting nutrient by activating the Keap1-Nrf2 system in different disease models. In addition, we wish to provide a more rational theoretical support for the medical applications of hydrogen.
Collapse
Affiliation(s)
- Danyu Cheng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (D.C.); (J.L.)
| | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (D.C.); (J.L.)
| | - Lin Zhao
- Cardiometabolic Innovation Center, Ministry of Education, Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (D.C.); (J.L.)
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
| |
Collapse
|
15
|
Tian J, Li Y, Mao X, Xie K, Zheng Y, Yu Y, Yu Y. Effects of the PI3K/Akt/HO-1 pathway on autophagy in a sepsis-induced acute lung injury mouse model. Int Immunopharmacol 2023; 124:111063. [PMID: 37857120 DOI: 10.1016/j.intimp.2023.111063] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/18/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
Sepsis-induced lung injury is an acute hypoxic respiratory insufficiency caused by systemic infectious factors that results in alveolar epithelial cell and capillary endothelial cell injury, diffuse pulmonary interstitial edema, and alveolar edema. Heme oxygenase (HO)-1 is usually associated with inflammation and has anti-inflammatory effects. Autophagy is a degradation pathway that eliminates cellular metabolic waste and plays an important protective role during stress. The phosphatidylinositol 3-kinase/ protein kinase B (PI3K/Akt) signaling pathway plays a key role in mediating cellular responses to inflammatory reactions. Therefore, we hypothesized that HO-1 is associated with autophagy and regulated by the PI3K/Akt signaling pathway in mice with sepsis-induced lung injury. Sepsis-induced lung injury was induced in mice using cecal ligation and puncture (CLP). Hemin or Sn-protoporphyrin IX (SnPP) was administered via intraperitoneal injection before surgery. Survival rates were observed during days 1-7 after the surgery; lung histology was discerned 24 h after the surgery; pro-inflammatory and anti-inflammatory factors in plasma and lung tissue were measured using enzyme-linked immunosorbent assay (ELISA); HO-1, Beclin-1, microtubule-associated protein 1 light chain 3B (LC3B)-II, p62 and lysosome associated membrane protein (LAMP)2 protein expression levels were measured 24 h after the surgery; HO-1 and LC3B-II protein expression levels were observed using immunofluorescence 24 h after the surgery; and autophagosomes were detected using electron microscopy 24 h after the surgery. Furthermore, when PI3K inhibitors LY294002, PI3K activators Recilisib and hemin were administered before the surgery, Akt, p-Akt, HO-1, and LC3-II levels were measured 24 h post-surgery. We found that HO-1 overexpression increased the survival rate and inhibited sepsis-induced lung injury. HO-1 overexpression attenuated the levels of proinflammatory cytokines (TNF-α, IL-1β) and increased the anti-inflammatory cytokine (IL-10, HO-1) overexpression. Moreover, HO-1 overexpression was also associated with increased expression of Beclin-1, LC3B-II and LAMP2 protein expression; decreased p62 protein expression; and significantly increased autophagosome formation. The results for HO-1-downregulated mice contrasted with those mentioned above. LY294002 inhibited p-Akt/Akt, HO-1, and LC3B-II protein expression; and hemin reversed the inhibitory effect of LY294002. The protective effect of HO-1 was involved in the mediation of autophagy, which may be regulated by the PI3K/Akt signaling pathway during sepsis-induced lung injury in mice.
Collapse
Affiliation(s)
- Jing Tian
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yanan Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Institute of Anesthesiology, Tianjin, China
| | - Xing Mao
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Institute of Anesthesiology, Tianjin, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yuxin Zheng
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Institute of Anesthesiology, Tianjin, China.
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China; Tianjin Institute of Anesthesiology, Tianjin, China.
| |
Collapse
|
16
|
Bai Y, Mi W, Meng X, Dong B, Jiang Y, Lu Y, Yu Y. Hydrogen alleviated cognitive impairment and blood‒brain barrier damage in sepsis-associated encephalopathy by regulating ABC efflux transporters in a PPARα-dependent manner. BMC Neurosci 2023; 24:37. [PMID: 37474902 PMCID: PMC10360271 DOI: 10.1186/s12868-023-00795-3] [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: 12/12/2022] [Accepted: 03/30/2023] [Indexed: 07/22/2023] Open
Abstract
Hydrogen (H2) can protect against blood‒brain barrier (BBB) damage in sepsis-associated encephalopathy (SAE), but the mechanism is still unclear. We examined whether it is related to PPARα and its regulatory targets, ABC efflux transporters. After injection with DMSO/GW6471 (a PPARα inhibitor), the mice subjected to sham/caecal ligation and puncture (CLP) surgery were treated with H2 for 60 min postoperation. Additionally, bEnd.3 cells were grown in DMSO/GW6471-containing or saline medium with LPS. In addition to the survival rates, cognitive function was assessed using the Y-maze and fear conditioning tests. Brain tissues were stained with TUNEL and Nissl staining. Additionally, inflammatory mediators (TNF-α, IL-6, HMGB1, and IL-1β) were evaluated with ELISA, and PPARα, ZO-1, occludin, VE-cadherin, P-gp, BCRP and MRP2 were detected using Western blotting. BBB destruction was assessed by brain water content and Evans blue (EB) extravasation. Finally, we found that H2 improved survival rates and brain dysfunction and decreased inflammatory cytokines. Furthermore, H2 decreased water content in the brain and EB extravasation and increased ZO-1, occludin, VE-cadherin and ABC efflux transporters regulated by PPARα. Thus, we concluded that H2 decreases BBB permeability to protect against brain dysfunction in sepsis; this effect is mediated by PPARα and its regulation of ABC efflux transporters.
Collapse
Affiliation(s)
- Yuanyuan Bai
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, PR China
| | - Wen Mi
- Department of Anesthesiology, Tianjin Baodi Hospital, Baodi Clinical College of Tianjin Medical University, Tianjin, 301800, China
| | - Xiaoyin Meng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Beibei Dong
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, PR China
| | - Yi Jiang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, PR China
| | - Yuechun Lu
- Department of Anesthesiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China.
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, PR China.
| |
Collapse
|
17
|
Htun Y, Nakamura S, Nakao Y, Mitsuie T, Ohta K, Arioka M, Yokota T, Inoue E, Inoue K, Tsuchiya T, Koyano K, Konishi Y, Miki T, Ueno M, Kusaka T. Conflicting findings on the effectiveness of hydrogen therapy for ameliorating vascular leakage in a 5-day post hypoxic-ischemic survival piglet model. Sci Rep 2023; 13:10486. [PMID: 37380745 DOI: 10.1038/s41598-023-37577-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 06/23/2023] [Indexed: 06/30/2023] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a major cause of morbidity and mortality in newborns in both high- and low-income countries. The important determinants of its pathophysiology are neural cells and vascular components. In neonatal HIE, increased vascular permeability due to damage to the blood-brain barrier is associated with seizures and poor outcomes in both translational and clinical studies. In our previous studies, hydrogen gas (H2) improved the neurological outcome of HIE and ameliorated the cell death. In this study, we used albumin immunohistochemistry to assess if H2 inhalation effectively reduced the cerebral vascular leakage. Of 33 piglets subjected to a hypoxic-ischemic insult, 26 piglets were ultimately analyzed. After the insult, the piglets were grouped into normothermia (NT), H2 ventilation (H2), therapeutic hypothermia (TH), and H2 combined with TH (H2-TH) groups. The ratio of albumin stained to unstained areas was analyzed and found to be lower in the H2 group than in the other groups, although the difference was not statistically significant. In this study, H2 therapy did not significantly improve albumin leakage despite the histological images suggesting signs of improvement. Further investigations are warranted to study the efficacy of H2 gas for vascular leakage in neonatal HIE.
Collapse
Affiliation(s)
- Yinmon Htun
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Yasuhiro Nakao
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Tsutomu Mitsuie
- Medical Engineering Equipment Management Center, Kagawa University Hospital, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Kenichi Ohta
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Makoto Arioka
- Maternal and Perinatal Center, Kagawa University Hospital, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Takayuki Yokota
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Eri Inoue
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Kota Inoue
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Toi Tsuchiya
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Kosuke Koyano
- Maternal and Perinatal Center, Kagawa University Hospital, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Yukihiko Konishi
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Takanori Miki
- Department of Anatomy and Neurobiology, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Masaki Ueno
- Department of Pathology and Host Defense, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, 1750-1 Mikicho, Kitagun, Kagawa, 761-0793, Japan.
| |
Collapse
|
18
|
Krzyzaniak K, Krion R, Szymczyk A, Stepniewska E, Sieminski M. Exploring Neuroprotective Agents for Sepsis-Associated Encephalopathy: A Comprehensive Review. Int J Mol Sci 2023; 24:10780. [PMID: 37445958 DOI: 10.3390/ijms241310780] [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: 05/30/2023] [Revised: 06/20/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Sepsis is a life-threatening condition resulting from an inflammatory overreaction that is induced by an infectious factor, which leads to multi-organ failure. Sepsis-associated encephalopathy (SAE) is a common complication of sepsis that can lead to acute cognitive and consciousness disorders, and no strict diagnostic criteria have been created for the complication thus far. The etiopathology of SAE is not fully understood, but plausible mechanisms include neuroinflammation, blood-brain barrier disruption, altered cerebral microcirculation, alterations in neurotransmission, changes in calcium homeostasis, and oxidative stress. SAE may also lead to long-term consequences such as dementia and post-traumatic stress disorder. This review aims to provide a comprehensive summary of substances with neuroprotective properties that have the potential to offer neuroprotection in the treatment of SAE. An extensive literature search was conducted, extracting 71 articles that cover a range of substances, including plant-derived drugs, peptides, monoclonal antibodies, and other commonly used drugs. This review may provide valuable insights for clinicians and researchers working in the field of sepsis and SAE and contribute to the development of new treatment options for this challenging condition.
Collapse
Affiliation(s)
- Klaudia Krzyzaniak
- Department of Emergency Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdansk, Poland
| | - Robert Krion
- Department of Emergency Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdansk, Poland
| | - Aleksandra Szymczyk
- Department of Emergency Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdansk, Poland
| | - Ewelina Stepniewska
- Department of Emergency Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdansk, Poland
| | - Mariusz Sieminski
- Department of Emergency Medicine, Medical University of Gdansk, Smoluchowskiego 17, 80-214 Gdansk, Poland
| |
Collapse
|
19
|
Zhou S, Li Y, Hong Y, Zhong Z, Zhao M. Puerarin protects against sepsis-associated encephalopathy by inhibiting NLRP3/Caspase-1/GSDMD pyroptosis pathway and reducing blood-brain barrier damage. Eur J Pharmacol 2023; 945:175616. [PMID: 36863556 DOI: 10.1016/j.ejphar.2023.175616] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/03/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023]
Abstract
Puerarin (Pue), an isoflavone compound extracted from Pueraria, has been shown to inhibit inflammation and reduce cerebral edema. The neuroprotective effect of puerarin has attracted much attention in recent years. Sepsis-associated encephalopathy (SAE) is a serious complication of sepsis that causes damage to the nervous system. This study aimed to investigate the effect of puerarin on SAE and elucidate the potential underlying mechanisms. A rat model of SAE was established by cecal ligation and puncture, and puerarin was injected intraperitoneally immediately after the operation. Puerarin was found to improve the survival rate and neurobehavioral score of SAE rats, alleviate symptoms, inhibit the level of brain injury markers NSE and S100β, and improve the pathological changes in rat brain tissue. Puerarin was also found to inhibit the level of factors related to the classical pathway of pyroptosis, such as NLRP3, Caspase-1, GSDMD, ASC, IL-1β, and IL-18. Puerarin also reduced the brain water content and penetration of Evan's Blue dye in SAE rats, and reduced the expression of MMP-9. In the in vitro experiments, we further confirmed the inhibitory effect of puerarin on neuronal pyroptosis by establishing a pyroptosis model in HT22 cells. Our findings suggest that puerarin may improve SAE by inhibiting the classical pathway of NLRP3/Caspase-1/GSDMD-mediated pyroptosis and reducing blood-brain barrier damage, thus playing a role in brain protection. Our study may provide a novel therapeutic strategy for SAE.
Collapse
Affiliation(s)
- Shuang Zhou
- Department of Emergency, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| | - Yuhua Li
- Department of Critical Care Medicine, Wuhan Children's Hospital, Wuhan, Hubei Province, 430014, China
| | - Yi Hong
- Department of Emergency, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Zhitao Zhong
- Department of Emergency, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China
| | - Min Zhao
- Department of Emergency, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, 110004, China.
| |
Collapse
|
20
|
Zhang Y, Chen J, Wu H, Li L, Yang X, Lai K, Bao J, Xie K, Yu Y. Hydrogen regulates mitochondrial quality to protect glial cells and alleviates sepsis-associated encephalopathy by Nrf2/YY1 complex promoting HO-1 expression. Int Immunopharmacol 2023; 118:110009. [PMID: 36963264 DOI: 10.1016/j.intimp.2023.110009] [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: 10/21/2022] [Revised: 02/20/2023] [Accepted: 03/06/2023] [Indexed: 03/26/2023]
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) is a complication of the central nervous system in patients with sepsis. Currently, no effective treatment for sepsis is available. Hydrogen plays a protective role in different diseases; however, the detailed mechanism of hydrogen-treated disease remains unclear. The purpose of this study was to investigate the effect of hydrogen on SAE in vitro and in vivo and the mechanism of hydrogen in mitochondrial dynamics and its function in astrocytes and microglia stimulated by lipopolysaccharides (LPSs). METHODS Animal models of SAE were generated by cecal ligation and puncture, and the SAE model was established by in vitro LPS stimulation. MTT, lactate dehydrogenase (LDH), reactive oxygen species (ROS), heme oxygenase-1 (HO-1) activity, mitochondrial membrane potential (MMP), and cell apoptosis assays were used to determine the effect of hydrogen on astrocytes and microglia stimulated by LPSs. The relationships between nuclear factor erythroid 2-related factor 2 (Nrf2), YY1, and HO-1 were examined by chromatin immunoprecipitation and co-immunoprecipitation. Mitochondrial homeostasis-related proteins in LPS-stimulated glial cells and brain tissues of SAE mice were detected by western blotting. The effects of hydrogen treatment in the SAE mouse model were investigated using Morris water maze and Y-maze analyses. RESULTS After performing experiments with different concentrations of LPSs in vitro, we selected 1000 ng/ml for subsequent experiments. Hydrogen attenuated the increase in ROS, LDH, and apoptosis and promoted decreases in cell activity and MMP, further promoting an increase in HO-1 expression induced by LPSs in astrocytes and microglia. Moreover, hydrogen further promoted the expression of Nrf2, HO-1, PGC-1α, TFAM, PARKIN, and PINK1, inhibited LPS-induced OPA1 and MFN2 expression in astrocytes and microglia, and downregulated the expression of DRP1 after LPS induction. Intriguingly, hydrogen treatment enhanced the binding between Nrf2 and YY1. However, silencing Nrf2 or YY1 abolished the protective effects of hydrogen on cell activity, LDH, ROS, and MMP; apoptosis; and regulation of Nrf2, HO-1, PGC-1α, TFAM, OPA1, DRP1, MFN2, PARKIN, and PINK1 in microglia. Finally, hydrogen treatment improved the results of behavioral detection, apoptosis, Nrf2, HO-1, PGC-1α, TFAM, OPA1, DRP1, MFN2, PARKIN, PINK1, and cytokines in SAE in vivo. CONCLUSIONS Hydrogen improved cell injury and mitochondrial quality, which were associated with HO-1 expression promoted by the Nrf2/YY1 complex in vitro. Thus, hydrogen treatment may represent a novel therapeutic method for treating SAE.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Anesthesiology, The Fourth Central Clinical School, Tianjin Medical University, Tianjin 300140, China; Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Juntai Chen
- Department of Anesthesiology, The Fourth Central Clinical School, Tianjin Medical University, Tianjin 300140, China
| | - Haidong Wu
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Lixin Li
- Good Laboratory Practice Center, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Xuejia Yang
- Good Laboratory Practice Center, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Keguan Lai
- Good Laboratory Practice Center, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Jingyu Bao
- Good Laboratory Practice Center, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China; Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| |
Collapse
|
21
|
Wang J, Yang S, Jing G, Wang Q, Zeng C, Song X, Li X. Inhibition of ferroptosis protects sepsis-associated encephalopathy. Cytokine 2023; 161:156078. [PMID: 36401983 DOI: 10.1016/j.cyto.2022.156078] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/17/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is a serious and common complication of sepsis. To study the ferroptosis in the pathogenesis of SAE and demonstrate the protection effect of ferroptosis resistance, cognitive function, neurological deficits, blood-brain barrier integrity and neuroinflammation were detected. SAE model was established by cecal ligation and puncture (CLP) in mice and an in vitro model was created by introducing LPS to HT22 cells. Ferroptosis inducer Fe-citrate (Fe) and ferroptosis inhibitor ferrostatin-1 (Fer-1) was post-treated in the models, respectively. SAE caused ferroptosis, as evidenced by an increase in reactive oxygen species (ROS), iron content and malondialdehyde (MDA) and a decrease in glutathione (GSH) level, as well as changes in the expression of ferroptosis-related proteins as acyl-CoA synthetase long-chain family member 4 (ACSL4), glutathione peroxidase 4 (GPX4), and cystine-glutamate antiporter (SLC7A11), and harmed mitochondrial function. In contrast, inhibiting ferroptosis with Fer-1 attenuated ferroptosis. Meanwhile, Fer-1 attenuated neurologic severity score, learning and memory impairment, Fluoro-Jade C (FJC) staining, and decreased Evans Blue (EB) extravasation, microglia activation and TNF-α and IL-1β production following SAE. The benefit of Fer-1 was diminished by ferroptosis inducer Fe. In addition, Fer-1 up-regulated the nuclear factor erythroid-2-related factor 2 (Nrf2)/ heme oxygenase-1(HO-1) signaling axis both in vivo and in vitro. In conclusion, our study revealed that Fer-1 might inhibit feroptosis in neurons by triggering the Nrf2/OH-1 pathway, thereby providing a therapeutic solution for SAE.
Collapse
Affiliation(s)
- Jin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shuhua Yang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guoqing Jing
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qingyuan Wang
- Department of Anesthesiology, the People's Hospital of Tuanfeng, Huanggang, Hubei, China
| | - Cheng Zeng
- Institute of Hepatobiliary Diseases of Wuhan University, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China; Transplant Center of Wuhan University, National Quality Control Center for Donated Organ Procurement, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, Hubei, China.
| | - Xuemin Song
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Xinyi Li
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
22
|
Yue J, Tan Y, Huan R, Guo J, Yang S, Deng M, Xiong Y, Han G, Liu L, Liu J, Cheng Y, Zha Y, Zhang J. Mast cell activation mediates blood-brain barrier impairment and cognitive dysfunction in septic mice in a histamine-dependent pathway. Front Immunol 2023; 14:1090288. [PMID: 36817492 PMCID: PMC9929573 DOI: 10.3389/fimmu.2023.1090288] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 01/13/2023] [Indexed: 02/04/2023] Open
Abstract
Introduction Sepsis-associated encephalopathy (SAE) is a diffuse cerebral dysfunction resulting from a systemic inflammatory response to infection; however, its pathophysiology remains unclear. Sepsis-induced neuroinflammation and blood-brain barrier (BBB) disruption are crucial factors in brain function disturbance in SAE. Mast cells (MCs) activation plays an important role in several neuroinflammation models; however, its role in SAE has not been comprehensively investigated. Methods We first established a SAE model by cecal ligation puncture (CLP) surgery and checked the activation of MCs. MCs activation was checked using immumohistochemical staining and Toluidine Blue staining. We administrated cromolyn (10mg/ml), a MC stabilizer, to rescue the septic mice. Brain cytokines levels were measured using biochemical assays. BBB disruption was assessed by measuring levels of key tight-junction (TJ) proteins. Cognitive function of mice was analyzed by Y maze and open field test. Transwell cultures of brain microvascular endothelial cells (BMVECs) co-cultured with MCs were used to assess the interaction of BMVECs and MCs. Results Results showed that MCs were overactivated in the hippocampus of CLP-induced SAE mice. Cromolyn intracerebroventricular (i.c.v) injection substantially inhibited the MCs activation and neuroinflammation responses, ameliorated BBB impairment, improved the survival rate and alleviated cognitive dysfunction in septic mice. In vitro experiments, we revealed that MCs activation increased the sensitivity of BMVECs against to lipopolysaccharide (LPS) challenge. Furthermore, we found that the histamine/histamine 1 receptor (H1R) mediated the interaction between MCs and BMVECs, and amplifies the LPS-induced inflammatory responses in BMVECs by modulating the TLR2/4-MAPK signaling pathway. Conclusions MCs activation could mediate BBB impairment and cognitive dysfunction in septic mice in a histamine-dependent pathway.
Collapse
Affiliation(s)
- Jianhe Yue
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Tan
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Renzheng Huan
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Guo
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Sha Yang
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Mei Deng
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yunbiao Xiong
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Guoqiang Han
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Lin Liu
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jian Liu
- Department of Neurosurgery, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Zha
- Department of Nephrology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Jiqin Zhang
- Department of Anesthesiology, Guizhou Provincial People's Hospital, Guiyang, China
| |
Collapse
|
23
|
Hydrogen Gas Treatment Improves Postoperative Delirium and Cognitive Dysfunction in Elderly Noncardiac Patients. J Pers Med 2022; 13:jpm13010067. [PMID: 36675728 PMCID: PMC9867387 DOI: 10.3390/jpm13010067] [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: 10/26/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/29/2022] Open
Abstract
Purpose: Postoperative delirium is a state of acute brain dysfunction characterized by fluctuating mental status that affects millions of patients each year. We used prophylactic inhalation of hydrogen gas in elderly patients undergoing elective surgery to compare their occurrence of postoperative delirium with that of controls. Methods: A total of 184 patients aged ≥ 65 years were enrolled and randomized into either a control group or a hydrogen inhalation group. The quality of sleep was assessed 1 day before and 1, 3, and 7 days after surgery at 8 A.M. The Confusion Assessment Method (CAM) was used as a screening tool for delirium and assessed the patients’ state of consciousness 1−7 days after surgery. Results: Postoperative delirium occurred in 17 (24%) of 70 patients without hydrogen inhalation and in 10 (12%) of 83 patients after hydrogen inhalation. The incidence of delirium was decreased in the hydrogen group. No significant differences were found between length of stay in hospital after surgery and sleep quality at 1, 3, and 7 days postoperatively between the two groups. The numerical rating scale (NRS) pain scores were higher in the hydrogen group (4.08 ± 1.77) than the control group (3.54 ± 1.77) on day 1 (p < 0.05); however, the mean difference between the two groups was small (1 to 1.6). There were no significant differences on day 3 and 7. The postoperative C-reactive protein level was significantly lower in the hydrogen group than the control group. Conclusions: This study suggests that hydrogen inhalation can prevent postoperative delirium in elderly noncardiac patients by reducing the inflammatory response.
Collapse
|
24
|
Electroacupuncture Alleviates Neuroinflammation by Inhibiting the HMGB1 Signaling Pathway in Rats with Sepsis-Associated Encephalopathy. Brain Sci 2022; 12:brainsci12121732. [PMID: 36552192 PMCID: PMC9776077 DOI: 10.3390/brainsci12121732] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/29/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Sepsis-Associated Encephalopathy (SAE) is common in sepsis patients, with high mortality rates. It is believed that neuroinflammation is an important mechanism involved in SAE. High mobility group box 1 protein (HMGB1), as a late pro-inflammatory factor, is significantly increased during sepsis in different brain regions, including the hippocampus. HMGB1 causes neuroinflammation and cognitive impairment through direct binding to advanced glycation end products (RAGE) and Toll-like receptor 4 (TLR4). Electroacupuncture (EA) at Baihui (GV20) and Zusanli (ST36) is beneficial for neurological diseases and experimental sepsis. Our study used EA to treat SAE induced by lipopolysaccharide (LPS) in male Sprague-Dawley rats. The Y maze test was performed to assess working memory. Immunofluorescence (IF) and Western blotting (WB) were used to determine neuroinflammation and the HMGB1 signaling pathway. Results showed that EA could improve working memory impairment in rats with SAE. EA alleviated neuroinflammation by downregulating the hippocampus's HMGB1/TLR4 and HMGB1/RAGE signaling, reducing the levels of pro-inflammatory factors, and relieving microglial and astrocyte activation. However, EA did not affect the tight junctions' expression of the blood-brain barrier (BBB) in the hippocampus.
Collapse
|
25
|
Zhao Q, Chen T, Ni C, Hu Y, Nan Y, Lin W, Liu Y, Zheng F, Shi X, Lin Z, Zhu J, Lin Z. Indole-3-propionic Acid Attenuates HI-Related Blood-Brain Barrier Injury in Neonatal Rats by Modulating the PXR Signaling Pathway. ACS Chem Neurosci 2022; 13:2897-2912. [PMID: 36129348 DOI: 10.1021/acschemneuro.2c00418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The blood-brain barrier (BBB) is an important physiological barrier of the human body contributing to maintaining brain homeostasis and normal function. Hypoxic-ischemic (HI)-related brain injury is one of the main causes of neonatal acute morbidity and chronic disability. The previous research of our group confirmed that there was serious BBB destruction during HI brain injury. However, at present, the protection strategy of BBB is very limited, and further research on the protection mechanism is warranted. Indole-3-propionic acid (IPA) is a bacterial metabolism with anti-inflammatory and antioxidant properties, having neuroprotective effects and protective effects on the mucosal barrier. However, the role of IPA in BBB is not clear. In this research, we demonstrated the protective effect of IPA on BBB disruption from HI brain injury and hypothesized that it involves the amelioration of inflammation, oxidative stress, and MMP activation, thereby inhibiting apoptosis of rat brain microvascular endothelial cells (rBMECs). We demonstrated that expression levels of several inflammatory markers, including iNOS, TNF-α, IL-6, and IL-1β, were significantly increased from HI damage or OGD injury. However, IPA treatment inhibited the increase significantly. Moreover, we demonstrated that IPA reduced intracellular ROS levels and MMP activation in rBMECs from OGD injury. Further research on the underlying detailed molecular mechanisms suggested that IPA attenuates inflammation by inhibiting NF-κB signaling. Finally, we investigated the mechanism of the relationship between PXR activation and NF-κB inhibition. The results suggested overexpression of PXR in rBMECs could significantly counteract the decrease of junction proteins and downregulate the increased p-IκB-α and p-NF-κB from OGD injury. However, the protective effects of IPA were reversed by antagonists of the PXR. Taken together, IPA might mitigate HI-induced damage of the BBB and the protective effect may be exerted through modulating the PXR signaling pathway.
Collapse
Affiliation(s)
- Qianlei Zhao
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Tingting Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Chao Ni
- Department of Pediatric Cardiovascular, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yingying Hu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yan Nan
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wei Lin
- Department of PICU, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yanli Liu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Feixia Zheng
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xulai Shi
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhongdong Lin
- Department of Pediatric Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jianghu Zhu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhenlang Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| |
Collapse
|
26
|
PPARα contributes to the therapeutic effect of hydrogen gas against sepsis-associated encephalopathy with the regulation to the CREB-BDNF signaling pathway and hippocampal neuron plasticity-related gene expression. Brain Res Bull 2022; 184:56-67. [DOI: 10.1016/j.brainresbull.2022.03.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 01/24/2023]
|
27
|
Li Y, Ji M, Yang J. Current Understanding of Long-Term Cognitive Impairment After Sepsis. Front Immunol 2022; 13:855006. [PMID: 35603184 PMCID: PMC9120941 DOI: 10.3389/fimmu.2022.855006] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Sepsis is recognized as a life-threatening multi-organ dysfunction resulting from a dysregulated host response to infection. Although the incidence and mortality of sepsis decrease significantly due to timely implementation of anti-infective and support therapies, accumulating evidence suggests that a great proportion of survivors suffer from long-term cognitive impairment after hospital discharge, leading to decreased life quality and substantial caregiving burdens for family members. Several mechanisms have been proposed for long-term cognitive impairment after sepsis, which are not mutually exclusive, including blood-brain barrier disruption, neuroinflammation, neurotransmitter dysfunction, and neuronal loss. Targeting these critical processes might be effective in preventing and treating long-term cognitive impairment. However, future in-depth studies are required to facilitate preventive and/or treatment strategies for long-term cognitive impairment after sepsis.
Collapse
Affiliation(s)
- Ying Li
- Department of Anesthesiology, Jiangyin Hospital, Affiliated to Southeast University Medical School, Jiangyin, China
| | - Muhuo Ji
- Department of Anesthesiology, The Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
Shi X, Zhu L, Wang S, Zhu W, Li Q, Wei J, Feng D, Liu M, Chen Y, Sun X, Lu H, Lv X. Magnesium Hydride Ameliorates Endotoxin-Induced Acute Respiratory Distress Syndrome by Inhibiting Inflammation, Oxidative Stress, and Cell Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5918954. [PMID: 35528515 PMCID: PMC9072031 DOI: 10.1155/2022/5918954] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 03/16/2022] [Accepted: 04/01/2022] [Indexed: 11/17/2022]
Abstract
Acute respiratory distress syndrome (ARDS) causes uncontrolled pulmonary inflammation, resulting in high morbidity and mortality in severe cases. Given the antioxidative effect of molecular hydrogen, some recent studies suggest the potential use of molecular hydrogen as a biomedicine for the treatment of ARDS. In this study, we aimed to explore the protective effects of magnesium hydride (MgH2) on two types of ARDS models and its underlying mechanism in a lipopolysaccharide (LPS)-induced ARDS model of the A549 cell line. The results showed that LPS successfully induced oxidative stress, inflammatory reaction, apoptosis, and barrier breakdown in alveolar epithelial cells (AEC). MgH2 can exert an anti-inflammatory effect by down-regulating the expressions of inflammatory cytokines (IL-1β, IL-6, and TNF-α). In addition, MgH2 decreased oxidative stress by eliminating intracellular ROS, inhibited apoptosis by regulating the expressions of cytochrome c, Bax, and Bcl-2, and suppressed barrier breakdown by up-regulating the expression of ZO-1 and occludin. Mechanistically, the expressions of p-AKT, p-mTOR, p-P65, NLRP3, and cleaved-caspase-1 were decreased after MgH2 treatment, indicating that AKT/mTOR and NF-κB/NLRP3/IL-1β pathways participated in the protective effects of MgH2. Furthermore, the in vivo study also demonstrated that MgH2-treated mice had a better survival rate and weaker pathological damage. All these findings demonstrated that MgH2 could exert an ARDS-protective effect by regulating the AKT/mTOR and NF-κB/NLRP3/IL-1β pathways to suppress LPS-induced inflammatory reaction, oxidative stress injury, apoptosis, and barrier breakdown, which may provide a potential strategy for the prevention and treatment of ARDS.
Collapse
Affiliation(s)
- Xuan Shi
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lina Zhu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Sheng Wang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wanli Zhu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Quanfu Li
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Juan Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Di Feng
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Meiyun Liu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanli Chen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuejun Sun
- Department of Naval Medicine, Naval Medical University, Shanghai, China
- Center of Hydrogen Science, Shanghai Jiao Tong University, Shanghai, China
| | - Hongtao Lu
- Department of Naval Medicine, Naval Medical University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
29
|
VIP alleviates sepsis-induced cognitive dysfunction as the TLR-4/NF-κB signaling pathway is inhibited in the hippocampus of rats. J Mol Histol 2022; 53:369-377. [PMID: 35239068 DOI: 10.1007/s10735-022-10068-8] [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] [Received: 08/13/2021] [Accepted: 12/29/2021] [Indexed: 01/17/2023]
Abstract
Cognitive dysfunction caused by sepsis-associated encephalopathy (SAE) is still poorly understood. It is reported that vasoactive intestinal peptide (VIP) exerts its anti-inflammatory effects in multiple diseases, while its biological function in SAE remains unclear. We aimed to figure out whether VIP has influence on sepsis-induced neuroinflammation and cognitive dysfunction. To induce sepsis, rats were subjected to cecal ligation and puncture (CLP) operation. Morris water maze test and fear conditioning test were conducted to reveal cognitive dysfunctions. TUNEL assay was performed to evaluate apoptosis. We found out that the expression of VIP was downregulated in the hippocampus of septic rats. VIP was verified to attenuate sepsis-induced memory impairment following CLP. Additionally, we examined apoptosis and inflammation in rats' hippocampus. It is worth noting that VIP inhibited the apoptosis in the hippocampus and reduced the productions of proinflammatory cytokines TNF-α, IL-6 and IL-1β. Furthermore, our data confirmed that VIP was involved in regulating the TLR-4/NF-κB signaling. In conclusion, VIP inhibited neuroinflammation and cognitive impairment in hippocampus of septic rats through the TLR-4/NF-κB signaling pathway.
Collapse
|
30
|
Zhang Y, Zhang J, Fu Z. Molecular hydrogen is a potential protective agent in the management of acute lung injury. Mol Med 2022; 28:27. [PMID: 35240982 PMCID: PMC8892414 DOI: 10.1186/s10020-022-00455-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome, which is a more severe form of ALI, are life-threatening clinical syndromes observed in critically ill patients. Treatment methods to alleviate the pathogenesis of ALI have improved to a great extent at present. Although the efficacy of these therapies is limited, their relevance has increased remarkably with the ongoing pandemic caused by the novel coronavirus disease 2019 (COVID-19), which causes severe respiratory distress syndrome. Several studies have demonstrated the preventive and therapeutic effects of molecular hydrogen in the various diseases. The biological effects of molecular hydrogen mainly involve anti-inflammation, antioxidation, and autophagy and cell death modulation. This review focuses on the potential therapeutic effects of molecular hydrogen on ALI and its underlying mechanisms and aims to provide a theoretical basis for the clinical treatment of ALI and COVID-19.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004 People’s Republic of China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004 People’s Republic of China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004 People’s Republic of China
| |
Collapse
|
31
|
Xiong Y, Yang J, Tong H, Zhu C, Pang Y. HMGB1 augments cognitive impairment in sepsis-associated encephalopathy by binding to MD-2 and promoting NLRP3-induced neuroinflammation. Psychogeriatrics 2022; 22:167-179. [PMID: 34931753 DOI: 10.1111/psyg.12794] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/07/2021] [Accepted: 11/23/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Sepsis-associated encephalopathy (SAE) always manifests with severe inflammatory symptoms and cognitive impairment. High mobility group box 1 (HMGB1) is a pro-inflammatory cytokine. In this study we investigated the role of HMGB1 in SAE. METHODS An SAE mouse model was established through cecal ligation and puncture surgery and then injected with adenovirus short hairpin RNA (Ad-sh)-HMGB1 or Ad-sh-myeloid differentiation protein (MD-2). The cognitive impairment and pathological injury in mice of different groups were evaluated using the Morris water maze experiment, Y-maze test, tail suspension test, fear conditioning test, and haematoxylin-eosin staining. The expressions of HMGB1 (fully reduced and disulfide (ds)HMGB1), MD-2, and NLRP3 in SAE mice were determined. Then, levels of inflammatory cytokines were measured. The binding relation between HMGB1 and MD-2 was predicted and certified. Additionally, MD-2 was downregulated to verify the role of the binding of HMGB1 and MD-2 in neuroinflammation and cognitive impairment in SAE. RESULTS Expressions of HMGB1, MD-2, NLRP3, and inflammatory cytokines were enhanced in the SAE mouse model, which were in parallel with impaired cognitive function. HMGB1 silencing resulted in downregulated NLRP3 expression and alleviated neuroinflammation and cognitive impairment in SAE mice. Mechanically, dsHMGB1 bound to MD-2 to activate NLRP3, thereby exacerbating neuroinflammation and cognitive impairment in SAE mice. The limited binding of HMGB1 and MD-2 downregulated NLRP3 expression to alleviate neuroinflammation and cognitive impairment in SAE mice. CONCLUSION HMGB1 was overexpressed in SAE, and dsHMGB1 bound to MD-2 to activate NLRP3 inflammasome, inducing neuroinflammation and cognitive impairment in SAE.
Collapse
Affiliation(s)
- Yanan Xiong
- Department of Emergency, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Jilin Yang
- Department of Emergency, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Haiyang Tong
- Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, P.R. China
| | - Chenting Zhu
- Department of Emergency, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| | - Yinhu Pang
- Department of Emergency, Shanghai Tongji Hospital, School of Medicine, Tongji University, Shanghai, P.R. China
| |
Collapse
|
32
|
Zhang Y, Chen S, Tian W, Zhu H, Li W, Dai W, Zhang X, Gu X, Su D. Emerging Trends and Hot Spots in Sepsis-Associated Encephalopathy Research From 2001 to 2021: A Bibliometric Analysis. Front Med (Lausanne) 2022; 9:817351. [PMID: 35295600 PMCID: PMC8918530 DOI: 10.3389/fmed.2022.817351] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/27/2022] [Indexed: 12/30/2022] Open
Abstract
Study Objectives To evaluate sepsis-associated encephalopathy (SAE) research and to quantitatively and qualitatively predict research hot spots using bibliometric analysis. Methods We extracted relevant publications from the Web of Science Core Collection on July 28, 2021. We investigated the retrieved data by bibliometric analysis (e.g. co-cited and cluster analysis, keyword co-occurrence) using the software CiteSpace and VOSviewer, the Online Analysis Platform of Literature Metrology (http://bibliometric.com/) and Bibliometrix to analyse and predict the trends and hot spots in this field. Main Results We identified 1,582 published articles and reviews on SAE from 2001 to 2021. During this period, the number of manuscripts on SAE increased steadily and peaked in 2021. The USA and China were the leading countries that had a critical impact on SAE research. Among all institutions, Vanderbilt University and Pittsburgh University held leading positions and became central in the collaboration network. Among all the journals, Critical Care Medicine published the maximum number of manuscripts in the field of SAE within 20 years. Dal-Pizzol Felipe was the most productive author (61 papers) and received the largest number of citations (930 citations). Co-citation cluster analysis revealed that the most popular terms on SAE in the manner of cluster labels were critical illness, sepsis-associated encephalopathy, polymicrobial sepsis, posterior reversible encephalopathy syndrome, rat brain, intensive care unit, prior sepsis, molecular hydrogen, inflammation drive, metabolic encephalopathies, delirium pathophysiology, and clinical neuroscience. Keyword burst detection indicated that neuroinflammation, blood-brain barrier (BBB) and mitochondria dysfunction were the current research hot spots. Conclusions Our study revealed that neuroinflammation, blood-brain barrier, and mitochondria dysfunction had been the research foci of SAE over the past 20 years. These have emerged as the basis for transformation from basic research to clinical application in finding effective methods for the prevention and treatment of SAE.
Collapse
|
33
|
Abstract
Molecular hydrogen exerts biological effects on nearly all organs. It has anti-oxidative, anti-inflammatory, and anti-aging effects and contributes to the regulation of autophagy and cell death. As the primary organ for gas exchange, the lungs are constantly exposed to various harmful environmental irritants. Short- or long-term exposure to these harmful substances often results in lung injury, causing respiratory and lung diseases. Acute and chronic respiratory diseases have high rates of morbidity and mortality and have become a major public health concern worldwide. For example, coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a global pandemic. An increasing number of studies have revealed that hydrogen may protect the lungs from diverse diseases, including acute lung injury, chronic obstructive pulmonary disease, asthma, lung cancer, pulmonary arterial hypertension, and pulmonary fibrosis. In this review, we highlight the multiple functions of hydrogen and the mechanisms underlying its protective effects in various lung diseases, with a focus on its roles in disease pathogenesis and clinical significance.
Collapse
Affiliation(s)
- Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jin Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| |
Collapse
|
34
|
Pu Y, Zhao L, Xi Y, Xia Y, Qian Y. The protective effects of Mirtazapine against lipopolysaccharide (LPS)-induced brain vascular hyperpermeability. Bioengineered 2022; 13:3680-3693. [PMID: 35081868 PMCID: PMC8973832 DOI: 10.1080/21655979.2021.2024962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sepsis is mainly characterized by severe inflammation triggered by infection, and sepsis-associated encephalopathy (SAE) is defined as brain damage caused by sepsis. Disruption of the blood-brain barrier (BBB) triggered by injured brain microvascular endothelial cells (BMECs) and damaged tight junction (TJ) structure is closely associated with the pathogenesis of SAE. The present research proposed to evaluate the potential therapeutic effects of Mirtazapine, a central presynaptic α2 receptor antagonist, on LPS-induced BBB disruption. The mice were administered with normal saline and 10 mg/kg Mirtazapine for 8 consecutive days, and from day 6, the experiment group of mice received LPS for 2 days to induce SAE. We found that the increased BBB permeability, elevated concentrations of inflammatory factors in brain tissues, and downregulated zonula occludens -1 (ZO-1) were observed in LPS-stimulated mice, all of which were reversed by 10 mg/kg Mirtazapine. In the in vitro assay, bEnd.3 brain endothelial cells were treated with 1 μM LPS in the absence or presence of Mirtazapine (25, 50 μM). We found that LPS-treated cells had significantly declined transendothelial electrical resistance (TEER), increased monolayer permeability, elevated production of inflammatory factors, and downregulated ZO-1. However, 25 and 50 μM Mirtazapine ameliorated all these LPS- induced aberrations. Mirtazapine also mitigated the decreased level of NF-E2-related factor 2 (Nrf2) in LPS-challenged endothelial cells. The protective effect of Mirtazapine on endothelial permeability against LPS was significantly abolished by the knockdown of Nrf2. Collectively, we concluded that Mirtazapine exerted protective effects on LPS-induced endothelial cells hyperpermeability by upregulating Nrf2.
Collapse
Affiliation(s)
- Yuehong Pu
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Zhao
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yao Xi
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yichun Xia
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Qian
- Department of Emergency Medicine, Yueyang Hospital of Intergrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
35
|
Chu J, Jiang Y, Zhou W, Zhang J, Li H, Yu Y, Yu Y. Acetaminophen alleviates ferroptosis in mice with sepsis-associated encephalopathy via the GPX4 pathway. Hum Exp Toxicol 2022; 41:9603271221133547. [DOI: 10.1177/09603271221133547] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is a cognitive impairment caused by sepsis, associated with increased morbidity and death. And acetaminophen (APAP) is a promising therapeutic medicine for SAE treatment. This research was designed to determine whether APAP alleviates SAE by attenuating ferroptosis and mediating the glutathione peroxidase (GPX4) pathway. The cecal ligation and puncture (CLP) approach was used to establish septic mouse models. The survival rates for 7 days were determined. The Morris water maze (MWM) was utilized to assess cognitive function. Hematoxylin and eosin (HE) staining identified histopathologic alterations in hippocampal tissue. Mitochondrial damage was discovered in hippocampal tissue using transmission electron microscopy (TEM). The reactive oxygen (ROS) levels in hippocampal tissue were measured using commercial assays. Septic cell models were produced using HT22 cells grown with 1 μg/ml lipopolysaccharide (LPS). ROS were quantified using immunofluorescence. Ferroptosis-related protein expression levels in hippocampal tissue and HT22 cells were measured using western blotting. To evaluate the iron content of hippocampal tissue and HT22 cells, commercial kits were employed. According to the findings, APAP improved survival rates, lowered hippocampal and mitochondrial damage, and improve cognitive impairment. In both animal and cell studies, APAP reduced iron content, ROS, glutamate antiporter (xCT), 4-hydroxy-2-nonenal (4-HNE) levels but increased GPX4 expression. However, RSL3, a GPX4 inhibitor that acts as a ferroptosis activator, decreased the protective properties of APAP in vitro. Our findings suggest that APAP reduces sepsis-induced cognitive impairment by reducing ferroptosis, which is mediated by the GPX4 signaling pathway.
Collapse
Affiliation(s)
- Jing Chu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
- Department of Anesthesiology, Specialized Medical Center of Chinese People’s Armed Police Force (PAP), Tianjin, China
| | - Yi Jiang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| | - Wenyu Zhou
- Department of Anesthesiology, Specialized Medical Center of Chinese People’s Armed Police Force (PAP), Tianjin, China
| | - Jialei Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| | - Hong Li
- Department of Anesthesiology, Specialized Medical Center of Chinese People’s Armed Police Force (PAP), Tianjin, China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Institute of Anesthesiology, Tianjin, China
| |
Collapse
|
36
|
Jeong ES, Bajgai J, You IS, Rahman MH, Fadriquela A, Sharma S, Kwon HU, Lee SY, Kim CS, Lee KJ. Therapeutic Effects of Hydrogen Gas Inhalation on Trimethyltin-Induced Neurotoxicity and Cognitive Impairment in the C57BL/6 Mice Model. Int J Mol Sci 2021; 22:ijms222413313. [PMID: 34948107 PMCID: PMC8703468 DOI: 10.3390/ijms222413313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/06/2021] [Accepted: 12/07/2021] [Indexed: 12/04/2022] Open
Abstract
Oxidative stress (OS) is one of the causative factors in the pathogenesis of various neurodegenerative diseases, including Alzheimer’s disease (AD) and cognitive dysfunction. In the present study, we investigated the effects of hydrogen (H2) gas inhalation in trimethyltin (TMT)-induced neurotoxicity and cognitive dysfunction in the C57BL/6 mice. First, mice were divided into the following groups: mice without TMT injection (NC), TMT-only injection group (TMT only), TMT injection + lithium chloride-treated group as a positive control (PC), and TMT injection + 2% H2 inhalation-treated group (H2). The TMT injection groups were administered a single dosage of intraperitoneal TMT injection (2.6 mg/kg body weight) and the H2 group was treated with 2% H2 for 30 min once a day for four weeks. Additionally, a behavioral test was performed with Y-maze to test the cognitive abilities of the mice. Furthermore, multiple OS- and AD-related biomarkers such as reactive oxygen species (ROS), nitric oxide (NO), calcium (Ca2+), malondialdehyde (MDA), glutathione peroxidase (GPx), catalase, inflammatory cytokines, apolipoprotein E (Apo-E), amyloid β (Aβ)-40, phospho-tau (p-tau), Bcl-2, and Bcl-2- associated X (Bax) were investigated in the blood and brain. Our results demonstrated that TMT exposure alters seizure and spatial recognition memory. However, after H2 treatment, memory deficits were ameliorated. H2 treatment also decreased AD-related biomarkers, such as Apo-E, Aβ-40, p-tau, and Bax and OS markers such as ROS, NO, Ca2+, and MDA in both serum and brain. In contrast, catalase and GPx activities were significantly increased in the TMT-only group and decreased after H2 gas treatment in serum and brain. In addition, inflammatory cytokines such as granulocyte colony-stimulating factors (G-CSF), interleukin (IL)-6, and tumor necrosis factor alpha (TNF-α) were found to be significantly decreased after H2 treatment in both serum and brain lysates. In contrast, Bcl-2 and vascular endothelial growth factor (VEGF) expression levels were found to be enhanced after H2 treatment. Taken together, our results demonstrated that 2% H2 gas inhalation in TMT-treated mice exhibits memory enhancing activity and decreases the AD, OS, and inflammatory-related markers. Therefore, H2 might be a candidate for repairing neurodegenerative diseases with cognitive dysfunction. However, further mechanistic studies are needed to fully clarify the effects of H2 inhalation on TMT-induced neurotoxicity and cognitive dysfunction.
Collapse
Affiliation(s)
- Eun-Sook Jeong
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea; (E.-S.J.); (J.B.); (M.H.R.); (S.S.); (C.-S.K.)
| | - Johny Bajgai
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea; (E.-S.J.); (J.B.); (M.H.R.); (S.S.); (C.-S.K.)
| | - In-Soo You
- GOOTZ Co., Ltd., 79-6, Yuljeong-ro 247 beon-gil, Yangju-si, Suwon 11457, Korea; (I.-S.Y.); (H.-U.K.); (S.-Y.L.)
| | - Md. Habibur Rahman
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea; (E.-S.J.); (J.B.); (M.H.R.); (S.S.); (C.-S.K.)
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
| | - Ailyn Fadriquela
- Department of Laboratory Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
| | - Subham Sharma
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea; (E.-S.J.); (J.B.); (M.H.R.); (S.S.); (C.-S.K.)
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
| | - Hwang-Un Kwon
- GOOTZ Co., Ltd., 79-6, Yuljeong-ro 247 beon-gil, Yangju-si, Suwon 11457, Korea; (I.-S.Y.); (H.-U.K.); (S.-Y.L.)
| | - So-Yeon Lee
- GOOTZ Co., Ltd., 79-6, Yuljeong-ro 247 beon-gil, Yangju-si, Suwon 11457, Korea; (I.-S.Y.); (H.-U.K.); (S.-Y.L.)
| | - Cheol-Su Kim
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea; (E.-S.J.); (J.B.); (M.H.R.); (S.S.); (C.-S.K.)
| | - Kyu-Jae Lee
- Department of Environmental Medical Biology, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea; (E.-S.J.); (J.B.); (M.H.R.); (S.S.); (C.-S.K.)
- Correspondence: ; Tel.: +82-(033)-741-331
| |
Collapse
|
37
|
Jiang B, Li Y, Dai W, Wu A, Wu H, Mao D. Hydrogen-rich saline alleviates early brain injury through regulating of ER stress and autophagy after experimental subarachnoid hemorrhage. Acta Cir Bras 2021; 36:e360804. [PMID: 34644772 PMCID: PMC8516430 DOI: 10.1590/acb360804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Subarachnoid hemorrhage (SAH) is a common complication of cerebral vascular disease. Hydrogen has been reported to alleviate early brain injury (EBI) through oxidative stress injury, reactive oxygen species (ROS), and autophagy. Autophagy is a programmed cell death mechanism that plays a vital role in neuronal cell death after SAH. However, the precise role of autophagy in hydrogen-mediated neuroprotection following SAH has not been confirmed. METHODS In the present study, the objective was to investigate the neuroprotective effects and potential molecular mechanisms of hydrogen-rich saline in SAH-induced EBI by regulating neural autophagy in the C57BL/6 mice model. Mortality, neurological score, brain water content, ROS, malondialdehyde (MDA), and neuronal death were evaluated. RESULTS The results show that hydrogen-rich saline treatment markedly increased the survival rate and neurological score, increased neuron survival, downregulated the autophagy protein expression of Beclin-1 and LC3, and endoplasmic reticulum (ER) stress. That indicates that hydrogen-rich saline-mediated inhibition of autophagy and ER stress ameliorate neuronal death after SAH. The neuroprotective capacity of hydrogen-rich saline is partly dependent on the ROS/Nrf2/heme oxygenase-1 (HO-1) signaling pathway. CONCLUSIONS The results of this study demonstrate that hydrogen-rich saline improves neurological outcomes in mice and reduces neuronal death by protecting against neural autophagy and ER stress.
Collapse
Affiliation(s)
| | | | | | - An Wu
- Wenzhou Medical University, China
| | | | | |
Collapse
|
38
|
Nie C, Zou R, Pan S, A R, Gao Y, Yang H, Bai J, Xi S, Wang X, Hong X, Yang W. Hydrogen gas inhalation ameliorates cardiac remodelling and fibrosis by regulating NLRP3 inflammasome in myocardial infarction rats. J Cell Mol Med 2021; 25:8997-9010. [PMID: 34402164 PMCID: PMC8435412 DOI: 10.1111/jcmm.16863] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/19/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
It is noteworthy that prolonged cardiac structural changes and excessive fibrosis caused by myocardial infarction (MI) seriously interfere with the treatment of heart failure in clinical practice. Currently, there are no effective and practical means of either prevention or treatment. Thus, novel therapeutic approaches are critical for the long‐term quality of life of individuals with myocardial ischaemia. Herein, we aimed to explore the protective effect of H2, a novel gas signal molecule with anti‐oxidative stress and anti‐inflammatory effects, on cardiac remodelling and fibrosis in MI rats, and to explore its possible mechanism. First, we successfully established MI model rats, which were then exposed to H2 inhalation with 2% concentration for 28 days (3 hours/day). The results showed that hydrogen gas can significantly improve cardiac function and reduce the area of cardiac fibrosis. In vitro experiments further proved that H2 can reduce the hypoxia‐induced damage to cardiomyocytes and alleviate angiotensin II‐induced migration and activation of cardiac fibroblasts. In conclusion, herein, we illustrated for the first time that inhalation of H2 ameliorates myocardial infarction‐induced cardiac remodelling and fibrosis in MI rats and exert its protective effect mainly through inhibiting NLRP3‐mediated pyroptosis.
Collapse
Affiliation(s)
- Chaoqun Nie
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rentong Zou
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuang Pan
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rong A
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC, Harbin Medical University, Harbin, China
| | - Yunan Gao
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongxiao Yang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Juncai Bai
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuiqing Xi
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue Wang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaojian Hong
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Yang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
39
|
Yu M, Qin C, Li P, Zhang Y, Wang Y, Zhang J, Li D, Wang H, Lu Y, Xie K, Yu Y, Yu Y. Hydrogen gas alleviates sepsis-induced neuroinflammation and cognitive impairment through regulation of DNMT1 and DNMT3a-mediated BDNF promoter IV methylation in mice. Int Immunopharmacol 2021; 95:107583. [PMID: 33773206 DOI: 10.1016/j.intimp.2021.107583] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/04/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
Sepsis-associated encephalopathy (SAE) can cause acute and long-term cognitive impairment and increase the mortality rate in sepsis patients, and we previously reported that 2% hydrogen gas (H2) inhalation has a therapeutic effect on SAE, but the underlying mechanism remains unclear. Dynamic DNA methylation, which catalyzed by DNA methyltransferases (DNMTs), is involved in the formation of synaptic plasticity and cognitive memory in the central nervous system. And brain-derived neurotrophic factor (BDNF), to be a key signaling component in activity-dependent synaptic plasticity, can be induced by neuronal activity accompanied by hypomethylation of its promoter IV. This study was designed to illustrate whether H2 can mediate SAE by alter the BDNF promoter IV methylation mediated by DNMTs. We established an SAE model by cecal ligation and perforation (CLP) in C57BL/6 mice. The Morris water maze test from the 4th to the 10th day after sham or CLP operations were used to evaluate mouse cognitive function. Hippocampal tissues were isolated at the 24 after sham or CLP surgery. Pro-inflammatory cytokines including tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6) and High Mobility Group Box 1 (HMGB1) were measured by enzyme-linked immunosorbent assay (ELISA). mRNA or protein levels of DNMTs (DNMT1, DNMT3a and DNMT3b), BDNF promoter IV and total BDNF were detected by RT-PCR and Western blot tests. Immunofluorescence staining were used to determine the expressions of DNMT1 and DNMT3a. The quantitative methylation analysis of the 11 CpG island of the promoter region of BDNF exon IV was determined using theAgena's MassARRAY EpiTYPER system. We found that 2% H2 inhalation can reduce pro-inflammatory factors, alleviate DNMT1, DNMT3a but not DNMT3b expression, make hypomethylation of BDNF promoter IV at 5 CpG sites, enhance the BDNF levels and then decrease escape latency but increase platform crossing times in septic mice. Our results suggest that 2% H2 inhalation may alleviate SAE through altering the regulation of BDNF promoter IV methylation which mediated by DNMT1 and DNMT3a in the hippocampus of septic mice.
Collapse
Affiliation(s)
- Mingdong Yu
- Department of Anesthesiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China; Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chao Qin
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Pei Li
- Department of Anesthesiology, Tianjin Hospital, Tianjin 300211, China
| | - Yingli Zhang
- Department of Anesthesiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Ying Wang
- Department of Anesthesiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Dedong Li
- Department of Anesthesiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Huixing Wang
- Pain Management Center, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yuechun Lu
- Department of Anesthesiology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| |
Collapse
|
40
|
Chen H, Dong B, Shi Y, Yu Y, Xie K. Hydrogen Alleviates Neuronal Injury and Neuroinflammation Induced by Microglial Activation via the Nuclear Factor Erythroid 2-related Factor 2 Pathway in Sepsis-associated Encephalopathy. Neuroscience 2021; 466:87-100. [PMID: 33992722 DOI: 10.1016/j.neuroscience.2021.05.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 04/30/2021] [Accepted: 05/04/2021] [Indexed: 12/17/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is characterized by diffuse cerebral and central nervous system (CNS) dysfunction. Microglia play a vital role in protecting the brain from neuronal damage, which is closely related to inflammatory responses. The nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway has an impact on microglial and neuronal injury. Here, we mainly explored the molecular mechanism by which Hydrogen (H2) regulates neuroinflammation in SAE and the role of Nrf2 in this process. An in vivo model of SAE was generated by cecal ligation and puncture (CLP). Primary microglia and neurons were cultured to establish an in vitro model. Microglia, neurons and brain tissue were obtained to detect Nrf2 expression, inflammation, cell injury, apoptosis, and microglial polarization. Escape latency, the number of platform crossings and the time spent in the target quadrant were measured to assess cognitive function. H2 attenuated microglial polarization from the M1 to the M2 phenotype, cytokine release and TLR/NF-κb activation and protected neurons from lipopolysaccharide (LPS)-activated microglia-induced injury via the Nrf2 pathway. SAE activated Nrf2 expression, and H2 further improved Nrf2 expression in SAE mice. H2 alleviated microglial polarization from the M1 to the M2 phenotype and cytokine release in the cerebral cortex and improved neuronal injury or cognitive dysfunction in SAE mice and wild-type mice but not in Nrf2-/- mice. H2 exerts antineuroinflammatory effects associated with TLR4/NF-κB signaling activation and neuroprotective effects by inhibiting the excessive release of proinflammatory cytokines, neuronal loss and apoptosis in vitro and in vivo through the Nrf2 pathway.
Collapse
Affiliation(s)
- Hongguang Chen
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Beibei Dong
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yuan Shi
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China; Tianjin Research Institute of Anesthesiology, Tianjin 300052, China.
| |
Collapse
|
41
|
Qi B, Yu Y, Wang Y, Wang Y, Yu Y, Xie K. Perspective of Molecular Hydrogen in the Treatment of Sepsis. Curr Pharm Des 2021; 27:667-678. [PMID: 32912119 DOI: 10.2174/1381612826666200909124936] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/09/2020] [Indexed: 11/22/2022]
Abstract
Sepsis is the main cause of death in critically ill patients with no effective treatment. Sepsis is lifethreatening organ dysfunction due to a dysregulated host response to infection. As a novel medical gas, molecular hydrogen (H2) has a therapeutic effect on many diseases, such as sepsis. H2 treatment exerts multiple biological effects, which can effectively improve multiple organ injuries caused by sepsis. However, the underlying molecular mechanisms of hydrogen involved in the treatment of sepsis remain elusive, which are likely related to anti-inflammation, anti-oxidation, anti-apoptosis, regulation of autophagy and multiple signaling pathways. This review can help better understand the progress of hydrogen in the treatment of sepsis, and provide a theoretical basis for the clinical application of hydrogen therapy in sepsis in the future.
Collapse
Affiliation(s)
- Bo Qi
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yaoqi Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yuzun Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
42
|
Li Q, Zhang H, Jia Z, Zhang L, Li Y, Xu R, Wang C, Yu Y. Hydrogen enriched saline alleviates morphine tolerance via inhibiting neuroinflammation, GLT-1, GS nitration and NMDA receptor trafficking and functioning in the spinal cord of rats. Neurosci Lett 2021; 755:135847. [PMID: 33774150 DOI: 10.1016/j.neulet.2021.135847] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 03/14/2021] [Accepted: 03/22/2021] [Indexed: 12/17/2022]
Abstract
The development and maintenance of morphine tolerance showed association with neuroinflammation and dysfunction of central glutamatergic system (such as nitration of glutamate transporter). Recent evidence indicated that hydrogen could reduce the levels of neuroinflammation and oxidative stress, but its role in morphine tolerance has not been studied. The rats were intrathecally administered with morphine (10 μg/10 μL each time, twice/day for 5 days). Hydrogen enriched saline (HS) or saline was given intraperitoneally at 1, 3 and 10 mL/kg for 10 min before each dose of morphine administration. The tail-flick latency, mechanical threshold and thermal latency were assessed one day (baseline) before and daily for up to 5 days during morphine injection. The pro-inflammatory cytokine expressions [tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), IL-6)] (by western blotting), astrocyte activation (by immunofluorescence and western blotting), and nitration of glutamate transporter-1 (GLT-1) and glutamine synthetase (GS) (by immunoprecipitation), membrane and total expression of N-methyl-d-aspartic acid (NMDA) receptor NR1 and NR2B subunits were carried out in the spinal dorsal horns. Chronic morphine administration induced antinociceptive tolerance, and together led to increased TNF-α, IL-1β and IL-6 expression, astrocyte activation, GLT-1 and GS nitration, increased membrane and total NR1, NR2B expression. Injection of HS attenuated morphine tolerance in a dose-dependent manner, decreased proinflammatory cytokine expression, inhibited astrocyte activation, decreased GLT-1 and GS nitration, and inhibited membrane trafficking of NMDA receptor. Our result showed that hydrogen pretreatment prevented morphine tolerance by reducing neuroinflammation, GLT-1, GS nitration, NMDA receptor trafficking in the spinal dorsal horn. Pretreatment with hydrogen might be considered as a novel therapeutic strategy for the prevention of morphine tolerance.
Collapse
Affiliation(s)
- Qing Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, PR China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, PR China.
| | - Haifang Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, PR China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, PR China.
| | - Zhen Jia
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, PR China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, PR China.
| | - Linlin Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, PR China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, PR China.
| | - Yize Li
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, PR China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, PR China.
| | - Rubin Xu
- Department of Anesthesiology, Tianjin First Central Hospital, Tianjin, 300192, PR China.
| | - Chunyan Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, PR China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, PR China.
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, PR China; Tianjin Research Institute of Anesthesiology, Tianjin, 300052, PR China.
| |
Collapse
|
43
|
Lian N, Shen M, Zhang K, Pan J, Jiang Y, Yu Y, Yu Y. Drinking Hydrogen-Rich Water Alleviates Chemotherapy-Induced Neuropathic Pain Through the Regulation of Gut Microbiota. J Pain Res 2021; 14:681-691. [PMID: 33732014 PMCID: PMC7956896 DOI: 10.2147/jpr.s288289] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
Introduction Chemotherapy-induced neuropathic pain (CINP) is one of the most common complications of chemotherapeutic drugs which limits the dose and duration of potentially life-saving anticancer treatment and compromises the quality of life of patients. Our previous studies have reported that molecular hydrogen (H2) can be used to prevent and treat various diseases. But the underlying mechanism remains unclear. The aim of the present study was to explore the effects of hydrogen-rich water on gut microbiota in CINP. Methods All C57BL/6J mice were divided into 4 groups: The group fed with normal drinking water and injected with saline (H2O + Saline), the group fed with normal drinking water and injected with oxaliplatin (H2O + OXA), the group fed with hydrogen-rich water and injected with saline (HW + Saline), and the group fed with hydrogen-rich water and injected with oxaliplatin (HW + OXA). The mechanical paw withdrawal threshold of the mice was tested on days 0, 5, 10, 15 and 20 after hydrogen-rich water treatment. On day 20, feces of mice from different groups were collected for microbial community diversity and structure analysis. The levels of inflammatory cytokines (TNF-α and IL-6), oxidative stress factors (OH- and ONOO-), lipopolysaccharide (LPS) and Toll-like receptor 4 (TLR4) were detected in dorsal root ganglia (DRG), L4-6 spinal cord segments and serum by enzyme-linked immunosorbent assay. The expression of TLR4 in DRG and spinal cords was determined by Western blot. Results The results illustrated that hydrogen-rich water could alleviate oxaliplatin-induced hyperalgesia, reduce the microbial diversity and alter the structure of gut microbiota, reverse the imbalance of inflammatory cytokines and oxidative stress, and decrease the expression of LPS and TLR4. Conclusion Hydrogen-rich water may alleviate CINP by affecting the diversity and structure of the gut microbiota, and then the LPS-TLR4 pathway, which provides a direction for further research.
Collapse
Affiliation(s)
- Naqi Lian
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Mengxi Shen
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Kai Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Jiacheng Pan
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Yi Jiang
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.,Tianjin Institute of Anesthesiology, Tianjin, People's Republic of China
| |
Collapse
|
44
|
Jiang Y, Zhang K, Yu Y, Wang Y, Lian N, Xie K, Yu Y. Molecular hydrogen alleviates brain injury and cognitive impairment in a chronic sequelae model of murine polymicrobial sepsis. Exp Brain Res 2020; 238:2897-2908. [PMID: 33052428 DOI: 10.1007/s00221-020-05950-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 10/06/2020] [Indexed: 01/13/2023]
Abstract
Sepsis-related encephalopathy (SAE), which causes a series of brain injuries and long-term, potentially irreversible cognitive dysfunction, is closely associated with increased morbidity and mortality. Hydrogen (H2) is a new type of medical gas molecule that has been widely used in the treatment of various diseases in recent years. The aim of the present study was to explore the protective effects of H2 inhalation on brain injury and long-term cognitive impairment in an improved chronic septic mouse model. Male C57BL/6J mice were randomized into four groups: Control, Control + H2, SAE and SAE + H2. The SAE and Control models were established by intraperitoneal injection of human stool suspension or saline in mice. H2 (2%) was inhaled for 60 min at 1 h and 6 h after SAE or Control treatment. The survival rates were recorded for 14 days (days 1-14) and the Morris Water Maze was performed for 7 days (days 8-14). To assess the severity of the brain injury, hematoxylin and eosin staining, Nissl staining, Evans blue (EB) extravasation and the wet/dry weight ratio of brain tissue were detected 24 h after SAE or Control treatment. In addition, inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin 6 (IL-6), high-mobility group box 1 (HMGB1), as well as the protein levels of nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), zonula occludens-1 (ZO-1) and Occludin, were measured 6, 12 and 24 h after SAE or Control treatment. The results showed that H2 treatment increased survival rates, mitigated cognitive impairment, reduced hippocampal histological damage, decreased EB and water content, and decreased the levels of TNF-α, IL-6, HMGB1, Nrf2, HO-1, ZO-1 and Occludin, as compared with the SAE group. These data revealed that 2% H2 could suppress brain damage and improve cognitive function in septic mice by inhibiting oxidative stress, inflammatory response and the sepsis-induced blood-brain barrier (BBB) disruption.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, People's Republic of China
- Tianjin Institute of Anesthesiology, Tianjin, 300052, People's Republic of China
| | - Kai Zhang
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, People's Republic of China
- Tianjin Institute of Anesthesiology, Tianjin, 300052, People's Republic of China
| | - Yang Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, People's Republic of China.
- Tianjin Institute of Anesthesiology, Tianjin, 300052, People's Republic of China.
| | - Yaoqi Wang
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, People's Republic of China
- Tianjin Institute of Anesthesiology, Tianjin, 300052, People's Republic of China
| | - Naqi Lian
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, People's Republic of China
- Tianjin Institute of Anesthesiology, Tianjin, 300052, People's Republic of China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, People's Republic of China
- Tianjin Institute of Anesthesiology, Tianjin, 300052, People's Republic of China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin, 300052, People's Republic of China.
- Tianjin Institute of Anesthesiology, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
45
|
Zhu JJ, Yu BY, Fu CC, He MZ, Zhu JH, Chen BW, Zheng YH, Chen SQ, Fu XQ, Li PJ, Lin ZL. LXA4 protects against hypoxic-ischemic damage in neonatal rats by reducing the inflammatory response via the IκB/NF-κB pathway. Int Immunopharmacol 2020; 89:107095. [PMID: 33096360 DOI: 10.1016/j.intimp.2020.107095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/23/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022]
Abstract
Hypoxia and the resultant decreases in cerebral blood flow in the perinatal period can lead to neonatal hypoxic-ischemic (HI) brain injury, which can, in turn, cause severe disability or even death. However, the efficacy of current treatment strategies remains limited. Several studies have demonstrated that lipoxin A4 (LXA4), as one of the earliest types of endogenous lipid mediators, can inhibit the accumulation of neutrophils, arrest inflammation, and promote the resolution of inflammation. However, research on LXA4 in the nervous system has rarely been carried out. In the present study, we sought to investigate the protective effect of LXA4 on HI brain damage in neonatal rats, as well as the underlying mechanisms. Through experiments conducted using an HI animal model, we found that the LXA4 intervention promoted the recovery of neuronal function and tissue structure following brain injury while maintaining the integrity of the blood-brain barrier in addition to reducing cerebral edema, infarct volume, and inflammatory responses. Our results suggest that LXA4 interfered with neuronal oxygen-glucose deprivation insults, reduced the expression of inflammatory factors, inhibited apoptosis, and promoted neuronal survival in vitro. Finally, the LXA4 intervention attenuated HI-induced activation of inhibitor kappa B (IκB) and degradation of nuclear factor-κB (NF-κB). In conclusion, our data suggest that LXA4 exerts a neuroprotective effect against neonatal HI brain damage through the IκB/NF-κB pathway. Our findings will help inform future studies regarding the effects of LXA4 on neuroinflammation, blood-brain barrier integrity, and neuronal apoptosis.
Collapse
Affiliation(s)
- Jin-Jin Zhu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Bin-Yuan Yu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Chang-Chang Fu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Min-Zhi He
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jiang-Hu Zhu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Bin-Wen Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yi-Hui Zheng
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shang-Qin Chen
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiao-Qin Fu
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Pei-Jun Li
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhen-Lang Lin
- Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China.
| |
Collapse
|