1
|
Pascal-Moussellard V, Alcaraz JP, Tanguy S, Salomez-Ihl C, Cinquin P, Boucher F, Boucher E. Molecular hydrogen as a potential mediator of the antitumor effect of inulin consumption. Sci Rep 2025; 15:11482. [PMID: 40181080 PMCID: PMC11968927 DOI: 10.1038/s41598-025-96346-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/27/2025] [Indexed: 04/05/2025] Open
Abstract
Inulin consumption and dihydrogen (H2) administration both exert antitumor effects on preclinical models as well as in clinical trials. As H2 is one of the major byproducts of inulin fermentation by bacterial species of the gut microbiota (GM), we hypothesized that H2 could mediate the antitumor effects of inulin. To provide evidence in favor of this hypothesis, we first determined the pattern of H2-exposure to which mice are subjected after inulin administration and developed an inhaled hydrogen therapy (H2T) protocol replicating this pattern. We then compared the effects on circulating immunity of a two-week daily inulin gavage with those of the corresponding H2T. We also compared the effects of inulin supplementation to those of the corresponding H2T on implanted melanoma growth and infiltration by T lymphocytes. Inulin and H2T induced a similar increase in circulating CD4+ and CD8+ T cells. In addition, both treatments similarly inhibited melanoma tumor growth. These results support a mechanism by which the H2 resulting from inulin fermentation by the GM diffuses across the intestinal barrier and stimulates the immunosurveillance responsible for the antitumor effect.
Collapse
Affiliation(s)
| | - Jean-Pierre Alcaraz
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, Grenoble, 38000, France
| | - Stéphane Tanguy
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, Grenoble, 38000, France
| | - Cordélia Salomez-Ihl
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, Grenoble, 38000, France
- Univ. Grenoble Alpes, CHU Grenoble Alpes, Inserm, Grenoble, CIC1406, 38000, France
| | - Philippe Cinquin
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, Grenoble, 38000, France
- Univ. Grenoble Alpes, CHU Grenoble Alpes, Inserm, Grenoble, CIC1406, 38000, France
| | - François Boucher
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, Grenoble, 38000, France.
- Bâtiment Jean Roget, Université Grenoble Alpes, TIMC UMR, 5525, Equipe PRETA, La Tronche, 38700, France.
| | - Emilie Boucher
- Univ. Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, TIMC, Grenoble, 38000, France
| |
Collapse
|
2
|
Sun J, Chen H, Xu X, Dou Y, Wu B, Zhang H, Shang S, Sun W. Effect of maternal cigarette smoke exposure on COPD progression in offspring mice. Reprod Toxicol 2024; 128:108646. [PMID: 38880403 DOI: 10.1016/j.reprotox.2024.108646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
OBJECTIVE To investigate the impact of maternal smoking on chronic obstructive pulmonary disease (COPD) progression in offspring. METHODS Using female C57BL/6 J mice, a maternal cigarette smoke exposure (CSE) model was established. Mice were exposed to cigarette smoke for 2 hours/day, 7 days/week, with a minimum 4-hour interval between exposures. Experimental groups included control (Con), pregnancy exposure (AS), pre-pregnancy exposure (SA), and pre-pregnancy + pregnancy exposure (SS). Lung function tests (Penh, PAU, TVb, EF50, Tr) were conducted on male offspring at 7 weeks. Histopathology, electron microscopy, and protein level changes were examined. RESULTS Lung function tests revealed significant impairments in Penh, PAU, TVb, EF50, and Tr in offspring across all exposure scenarios. Specifically, AS experienced significant lung function impairment and mitochondrial dysfunction in offspring, with noticeable pulmonary lesions and increased apoptosis. SA showed similar or even more severe lung function impairment and cellular apoptosis. SS exhibited the most pronounced effects, with the highest levels of lung dysfunction, mitochondrial damage, and apoptosis. Histopathological analysis showed pulmonary lesions in offspring exposed to maternal CSE. Flow cytometry revealed increased apoptosis and reduced mitochondrial membrane potential in offspring lung cells. Electron microscopy confirmed mitochondrial dysfunction. Upregulation of apoptotic proteins and downregulation of anti-apoptotic protein Bcl-2 were found in offspring lung tissue exposed to maternal CSE. CONCLUSION Maternal smoking induces impaired lung function, pulmonary lesions, and mitochondrial dysfunction in offspring, regardless of exposure timing and duration. Additionally, it alters expression of apoptosis-related proteins in offspring lung tissue, potentially contributing to COPD susceptibility.
Collapse
Affiliation(s)
- Jiawei Sun
- Department of Respiratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang 050031, China
| | - Huan Chen
- Department of Anesthesiology, The First Hospital of Hebei Medical University, Shijiazhuang 050031, China
| | - Xu Xu
- Department of Respiratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang 050031, China
| | - Yaping Dou
- Department of Respiratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang 050031, China
| | - Baofa Wu
- Department of Respiratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang 050031, China
| | - Hongyang Zhang
- Department of Respiratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang 050031, China
| | - Song Shang
- Department of Respiratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang 050031, China
| | - Wuzhuang Sun
- Department of Respiratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang 050031, China.
| |
Collapse
|
3
|
Zhang C, Xing Y, Wu X, Jiang Q, Luo X, He W, Liu S, Lu W, Wang J. Inhalation of hydrogen gas protects against mitomycin-induced pulmonary veno-occlusive disease. Respir Res 2024; 25:281. [PMID: 39014440 PMCID: PMC11253336 DOI: 10.1186/s12931-024-02906-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 07/04/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND As a subtype of pulmonary hypertension (PH), pulmonary veno-occlusive disease (PVOD) is devastating and life-threatening disease without effective therapy. Hydrogen has been reported to exhibits antioxidant and anti-inflammatory effects in a rat model induced by monocrotaline of PH. In this study, we investigated the effects of inhaled hydrogen gas on the prevention and treatment of PVOD induced by mitomycin C (MMC) in rats. METHODS PVOD was induced in female Sprague-Dawley rats through intraperitoneal injection of MMC at a concentration of 3 mg·kg- 1·wk- 1 for 2 weeks. Inhalation of hydrogen gas (H2) was administered through a designed rat cage concurrently or two weeks after MMC administration. The severity of PVOD was assessed by using hemodynamic measurements and histological analysis. The expression levels of general control nonderepressible 2 (GCN2), nuclear factor erythroid 2-related factor-2 (Nrf2), heme oxygenase-1 (HO-1) and endothelial-to-mesenchymal transition (EndoMT) related proteins in lung tissue were measured. Levels of lipid peroxidation pro-inflammatory cytokines in serum were determined. RESULTS Inhaled H2 improved hemodynamics and right heart function, reversed right ventricular hypertrophy, and prevented pulmonary vessel reconstitution in both prevention and treatment approaches. It decreased malondialdehyde (MDA) levels in the serum and the expression of NADPH oxidase 1 (NOX-1) in lung tissue. It regulated Nrf2/HO-1 signaling pathway and anti-inflammatory factor GCN2 in lung tissue, accompanied by a decrease in macrophages and pro-inflammatory cytokines. Our data suggested that H2 inhalation effectively countered EndoMT induced by MMC, as evidenced by the detection of endothelial markers (e.g., VE-cadherin and CD31) and mesenchymal markers (e.g., vimentin and fibronectin). Further research revealed that H2 preserved p-Smad3 and induced p-Smad1/5/9. CONCLUSION Inhalation of H2 effectively inhibits the pathogenesis of PVOD induced by MMC in rats. This inhibitory effect may be attributed to the antioxidant and anti-inflammatory properties of H2.
Collapse
Affiliation(s)
- Chenting Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Yue Xing
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Xuefen Wu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Qian Jiang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Xiaoyun Luo
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Wei He
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Shiyun Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Wenju Lu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China.
- Section of Physiology, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA.
| |
Collapse
|
4
|
Gao X, Niu S, Li L, Zhang X, Cao X, Zhang X, Pan W, Sun M, Zhao G, Zheng X, Song G, Zhang Y. Hydrogen therapy promotes macrophage polarization to the M2 subtype in radiation lung injury by inhibiting the NF-κB signalling pathway. Heliyon 2024; 10:e30902. [PMID: 38826750 PMCID: PMC11141264 DOI: 10.1016/j.heliyon.2024.e30902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 06/04/2024] Open
Abstract
Background Radiotherapy has become a standard treatment for chest tumors, but a common complication of radiotherapy is radiation lung injury. Currently, there is still a lack of effective treatment for radiation lung injury. Methods A mouse model of radioactive lung injury (RILI) was constructed and then treated with different cycles of hydrogen inhalation. Lung function tests were performed to detect changes in lung function.HE staining was used to detect pathological changes in lung tissue. Immunofluorescence staining was used to detect the polarization of macrophages in lung tissue. Immunohistochemistry was used to detect changes in cytokine expression in lung tissues. Western Blot was used to detect the expression of proteins related to the NF-κB signalling pathway. Results Lung function test results showed that lung function decreased in the model group and improved in the treatment group.HE staining showed that inflammatory response was evident in the model group and decreased in the treatment group. Immunohistochemistry results showed that the expression of pro-inflammatory factors was significantly higher in the model group, and the expression of pro-inflammatory factors was significantly higher in the treatment group. The expression of pro-inflammatory factors in the treatment group was significantly lower than that in the model group, and the expression of anti-inflammatory factors in the treatment group was higher than that in the model group. Immunofluorescence showed that the expression of M1 subtype macrophages was up-regulated in the model group and down-regulated in the treatment group. The expression of M2 subtype macrophages was up-regulated in the treatment group relative to the model group. Western Blot showed that P-NF-κB p65/NF-κB p65 was significantly increased in the model group, and P-NF-κB p65/NF-κB p65 was decreased in the treatment group. Conclusion Hydrogen therapy promotes macrophage polarization from M1 to M2 subtypes by inhibiting the NF-κB signalling pathway, thereby attenuating the inflammatory response to radiation lung injury.
Collapse
Affiliation(s)
- Xue Gao
- Department of Pathophysiology, School of Clinical and Basic Medicine, Shandong First Medical University, China
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Shiying Niu
- Department of Pathophysiology, School of Clinical and Basic Medicine, Shandong First Medical University, China
- Department of Pathology, Linfen Central Hospital, China
| | - Lulu Li
- Department of Pathophysiology, School of Clinical and Basic Medicine, Shandong First Medical University, China
| | - Xiaoyue Zhang
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Xuetao Cao
- Department of Pathophysiology, School of Clinical and Basic Medicine, Shandong First Medical University, China
| | - Xinhui Zhang
- Department of Pathophysiology, School of Clinical and Basic Medicine, Shandong First Medical University, China
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Wentao Pan
- Department of Pathophysiology, School of Clinical and Basic Medicine, Shandong First Medical University, China
| | - Meili Sun
- Department of Pathology, Linfen Central Hospital, China
- Department of Oncology, Affiliated Central Hospital of Shandong First Medical University, China
| | - Guoli Zhao
- Department of Pathology, Liaocheng Infectious Disease Hospital, China
| | - Xuezhen Zheng
- Department of Pathophysiology, School of Clinical and Basic Medicine, Shandong First Medical University, China
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, China
| | - Guohua Song
- Department of Pathophysiology, School of Clinical and Basic Medicine, Shandong First Medical University, China
| | - Yueying Zhang
- Department of Pathophysiology, School of Clinical and Basic Medicine, Shandong First Medical University, China
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, China
| |
Collapse
|
5
|
Akaras N, Ileriturk M, Gur C, Kucukler S, Oz M, Kandemir FM. The protective effects of chrysin on cadmium-induced pulmonary toxicity; a multi-biomarker approach. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:89479-89494. [PMID: 37453011 DOI: 10.1007/s11356-023-28747-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
This study aimed to determine the potential protective effects of chrysin (CHR) on experimental cadmium (Cd)-induced lung toxicity in rats. To this end, rats were divided into five groups; Control, CHR, Cd, Cd + CHR25, Cd + CHR50. In the study, rats were treated with CHR (oral gavage, 25 mg/kg and 50 mg/kg) 30 min after giving Cd (oral gavage, 25 mg/kg) for 7 consecutive days. The effects of Cd and CHR treatments on oxidative stress, inflammatory response, ER stress, apoptosis and tissue damage in rat lung tissues were determined by biochemical and histological methods. Our results revealed that CHR therapy for Cd-administered rats could significantly reduce MDA levels in lung tissue while significantly increasing the activity of antioxidant enzymes (SOD, CAT, GPx) and GSH levels. CHR agent exerted antiinflammatory effect by lowering elevated levels of NF-κB, IL-1β IL-6, TNF-α, RAGE and NRLP3 in Cd-induced lung tissue. Moreover CHR down-regulated Cd-induced ER stress markers (PERK, IRE1, ATF6, CHOP, and GRP78) and apoptosis markers (Caspase-3, Bax) lung tissue. CHR up-regulated the Bcl-2 gene, an anti-apoptotic marker. Besides, CHR attenuated the side effects caused by Cd by modulating histopathological changes such as hemorrhage, inflammatory cell infiltration, thickening of the alveolar wall and collagen increase. Immunohistochemically, NF-κB and Caspase-3 expressions were intense in the Cd group, while these expressions were decreased in the Cd + CHR groups. These results suggest that CHR exhibits protective effects against Cd-induced lung toxicity in rats by ameliorating oxidative stress, inflammation, apoptosis, endoplasmic reticulum stress and histological changes.
Collapse
Affiliation(s)
- Nurhan Akaras
- Department of Histology and Embryology, Faculty of Medicine, Aksaray University, Aksaray, Turkey.
| | - Mustafa Ileriturk
- Department of Animal Science, Horasan Vocational College, Atatürk University, Erzurum, Turkey
| | - Cihan Gur
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Sefa Kucukler
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Mehmet Oz
- Department of Physiology, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| | - Fatih Mehmet Kandemir
- Department of Medical Biochemistry, Faculty of Medicine, Aksaray University, Aksaray, Turkey
| |
Collapse
|
6
|
Perveen I, Bukhari B, Najeeb M, Nazir S, Faridi TA, Farooq M, Ahmad QUA, Abusalah MAHA, ALjaraedah TY, Alraei WY, Rabaan AA, Singh KKB, Abusalah MAHA. Hydrogen Therapy and Its Future Prospects for Ameliorating COVID-19: Clinical Applications, Efficacy, and Modality. Biomedicines 2023; 11:1892. [PMID: 37509530 PMCID: PMC10377251 DOI: 10.3390/biomedicines11071892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 07/30/2023] Open
Abstract
Molecular hydrogen is renowned as an odorless and colorless gas. The recommendations developed by China suggest that the inhalation of hydrogen molecules is currently advised in COVID-19 pneumonia treatment. The therapeutic effects of molecular hydrogens have been confirmed after numerous clinical trials and animal-model-based experiments, which have expounded that the low molecular weight of hydrogen enables it to easily diffuse and permeate through the cell membranes to produce a variety of biological impacts. A wide range of both chronic and acute inflammatory diseases, which may include sepsis, pancreatitis, respiratory disorders, autoimmune diseases, ischemia-reperfusion damages, etc. may be treated and prevented by using it. H2 can primarily be inoculated through inhalation, by drinking water (which already contains H2), or by administrating the injection of saline H2 in the body. It may play a pivotal role as an antioxidant, in regulating the immune system, in anti-inflammatory activities (mitochondrial energy metabolism), and cell death (apoptosis, pyroptosis, and autophagy) by reducing the formation of excessive reactive O2 species and modifying the transcription factors in the nuclei of the cells. However, the fundamental process of molecular hydrogen is still not entirely understood. Molecular hydrogen H2 has a promising future in therapeutics based on its safety and possible usefulness. The current review emphasizes the antioxidative, anti-apoptotic, and anti-inflammatory effects of hydrogen molecules along with the underlying principle and fundamental mechanism involved, with a prime focus on the coronavirus disease of 2019 (COVID-19). This review will also provide strategies and recommendations for the therapeutic and medicinal applications of the hydrogen molecule.
Collapse
Affiliation(s)
- Ishrat Perveen
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore 54590, Pakistan
| | - Bakhtawar Bukhari
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore 54590, Pakistan
| | - Mahwish Najeeb
- University Institute of Public Health, The University of Lahore, Lahore 54590, Pakistan
| | - Sumbal Nazir
- School of Zoology, Minhaj University Lahore, Lahore 54770, Pakistan
| | - Tallat Anwar Faridi
- University Institute of Public Health, The University of Lahore, Lahore 54590, Pakistan
| | - Muhammad Farooq
- Food and Biotechnology Research Centre, Pakistan Council of Scientific and Industrial Research Centre, Lahore 54590, Pakistan
| | - Qurat-Ul-Ain Ahmad
- Division of Science and Technology, University of Education, Township Lahore, Lahore 54770, Pakistan
| | - Manal Abdel Haleem A Abusalah
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Thana' Y ALjaraedah
- Department of Diet Therapy Technology & Dietetics, Faculty of Allied Medical Sciences, Zarqa University, Al-Zarqa 13132, Jordan
| | - Wesal Yousef Alraei
- Department of Diet Therapy Technology & Dietetics, Faculty of Allied Medical Sciences, Zarqa University, Al-Zarqa 13132, Jordan
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
| | - Kirnpal Kaur Banga Singh
- Department of Medical Microbiology and Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Mai Abdel Haleem A Abusalah
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Zarqa University, Al-Zarqa 13132, Jordan
| |
Collapse
|
7
|
Artamonov MY, Martusevich AK, Pyatakovich FA, Minenko IA, Dlin SV, LeBaron TW. Molecular Hydrogen: From Molecular Effects to Stem Cells Management and Tissue Regeneration. Antioxidants (Basel) 2023; 12:antiox12030636. [PMID: 36978884 PMCID: PMC10045005 DOI: 10.3390/antiox12030636] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
It is known that molecular hydrogen is a relatively stable, ubiquitous gas that is a minor component of the atmosphere. At the same time, in recent decades molecular hydrogen has been shown to have diverse biological effects. By the end of 2022, more than 2000 articles have been published in the field of hydrogen medicine, many of which are original studies. Despite the existence of several review articles on the biology of molecular hydrogen, many aspects of the research direction remain unsystematic. Therefore, the purpose of this review was to systematize ideas about the nature, characteristics, and mechanisms of the influence of molecular hydrogen on various types of cells, including stem cells. The historical aspects of the discovery of the biological activity of molecular hydrogen are presented. The ways of administering molecular hydrogen into the body are described. The molecular, cellular, tissue, and systemic effects of hydrogen are also reviewed. Specifically, the effect of hydrogen on various types of cells, including stem cells, is addressed. The existing literature indicates that the molecular and cellular effects of hydrogen qualify it to be a potentially effective agent in regenerative medicine.
Collapse
Affiliation(s)
- Mikhail Yu. Artamonov
- Laboratory of Translational Free Radical Biomedicine, Sechenov University, 119991 Moscow, Russia
- MJA Research and Development, Inc., East Stroudsburg, PA 18301, USA
- Correspondence: (M.Y.A.); (T.W.L.); Tel.: +1-570-972-6778 (M.Y.A.); +1-435-586-7818 (T.W.L.)
| | - Andrew K. Martusevich
- Laboratory of Translational Free Radical Biomedicine, Sechenov University, 119991 Moscow, Russia
- Laboratory of Medical Biophysics, Privolzhsky Research Medical University, 603005 Nizhny Novgorod, Russia
| | | | - Inessa A. Minenko
- Laboratory of Translational Free Radical Biomedicine, Sechenov University, 119991 Moscow, Russia
- MJA Research and Development, Inc., East Stroudsburg, PA 18301, USA
| | - Sergei V. Dlin
- MJA Research and Development, Inc., East Stroudsburg, PA 18301, USA
| | - Tyler W. LeBaron
- Department of Kinesiology and Outdoor Recreation, Southern Utah University, Cedar City, UT 84720, USA
- Molecular Hydrogen Institute, Enoch, UT 84721, USA
- Correspondence: (M.Y.A.); (T.W.L.); Tel.: +1-570-972-6778 (M.Y.A.); +1-435-586-7818 (T.W.L.)
| |
Collapse
|
8
|
Yang HJ, Tsou WH, Shen MC, Liu CY, Saunders HM, Wang KY, Douglas FL. The effects of hydrogen treatment in a cigarette smoke solution-induced chronic obstructive pulmonary disease-like changes in an animal model. J Thorac Dis 2022; 14:4246-4255. [PMID: 36524091 PMCID: PMC9745525 DOI: 10.21037/jtd-22-324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022]
Abstract
Background Molecular hydrogen, with its antioxidant and anti-inflammatory properties, may be suitable for the prevention and treatment of chronic obstructive pulmonary disease (COPD). This study aims to investigate the therapeutic efficacy of hydrogen-oxygen (H2/O2) treatment in cigarette smoke solution (CSS)-induced COPD-like injury in a female BALB/c mouse model. Methods Thirty mice were randomly assigned to three groups: Control (n=8), COPD (n=10), and COPD + H2/O2 (n=12). CSS was administered by intraperitoneal (IP) injection twice weekly for 6 weeks during the COPD induction phase. Simultaneously, the COPD + H2/O2 group started received 75 minutes of inhalation therapy (42% H2) delivered by the Oxy-Hydrogen Generator twice daily for 9 weeks. Mice body weights and survival were measured throughout the study period. Neutrophil elastase (NE) activity and lung histopathological changes were also evaluated. Results The results showed a higher survival rate in the COPD + H2/O2 group compared to the COPD group (100% vs. 80%) during the induction phase. Slight decreases in body weight gains were observed in the COPD and COPD + H2/O2 groups during the first 15 days of the induction phase, but there was no significant difference in mean body weights among the three groups throughout the study period. NE activity was numerically lower in the COPD + H2/O2 group compared to the COPD group. The histopathological evaluation showed significant improvements in the H2/O2-treated mice with respect to mean linear intercept (MLI) and lesion (inflammation and emphysema) scores. Improvements in goblet cell hypertrophy and hyperplasia of airway epithelium were not significant. Conclusions A 9-week H2/O2 inhalation therapy delivered by the Oxy-Hydrogen Generator to CSS-induced COPD-like injury in mice showed improvement in survival rate, alveolar structural changes, and histopathological lesion scores of the lung.
Collapse
Affiliation(s)
| | | | | | | | - Hsiu-Ming Saunders
- HOHO Biotech Co., Ltd., Taipei.,IPC Intellectual Property Connections, Inc., Palo Alto, CA, USA
| | | | | |
Collapse
|
9
|
Aghapour M, Ubags ND, Bruder D, Hiemstra PS, Sidhaye V, Rezaee F, Heijink IH. Role of air pollutants in airway epithelial barrier dysfunction in asthma and COPD. Eur Respir Rev 2022; 31:31/163/210112. [PMID: 35321933 PMCID: PMC9128841 DOI: 10.1183/16000617.0112-2021] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 11/13/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic exposure to environmental pollutants is a major contributor to the development and progression of obstructive airway diseases, including asthma and COPD. Understanding the mechanisms underlying the development of obstructive lung diseases upon exposure to inhaled pollutants will lead to novel insights into the pathogenesis, prevention and treatment of these diseases. The respiratory epithelial lining forms a robust physicochemical barrier protecting the body from inhaled toxic particles and pathogens. Inhalation of airborne particles and gases may impair airway epithelial barrier function and subsequently lead to exaggerated inflammatory responses and airway remodelling, which are key features of asthma and COPD. In addition, air pollutant-induced airway epithelial barrier dysfunction may increase susceptibility to respiratory infections, thereby increasing the risk of exacerbations and thus triggering further inflammation. In this review, we discuss the molecular and immunological mechanisms involved in physical barrier disruption induced by major airborne pollutants and outline their implications in the pathogenesis of asthma and COPD. We further discuss the link between these pollutants and changes in the lung microbiome as a potential factor for aggravating airway diseases. Understanding these mechanisms may lead to identification of novel targets for therapeutic intervention to restore airway epithelial integrity in asthma and COPD. Exposure to air pollution induces airway epithelial barrier dysfunction through several mechanisms including increased oxidative stress, exaggerated cytokine responses and impaired host defence, which contributes to development of asthma and COPD. https://bit.ly/3DHL1CA
Collapse
Affiliation(s)
- Mahyar Aghapour
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Niki D Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, CHUV, Epalinges, Switzerland
| | - Dunja Bruder
- Infection Immunology Group, Institute of Medical Microbiology, Infection Control and Prevention, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany.,Immune Regulation Group, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Pieter S Hiemstra
- Dept of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Venkataramana Sidhaye
- Pulmonary and Critical Care Medicine, Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Fariba Rezaee
- Center for Pediatric Pulmonary Medicine, Cleveland Clinic Children's, Cleveland, OH, USA.,Dept of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Irene H Heijink
- University of Groningen, University Medical Center Groningen, Depts of Pathology and Medical Biology and Pulmonology, Groningen Research Institute for Asthma and COPD (GRIAC), Groningen, The Netherlands
| |
Collapse
|
10
|
Role of Molecular Hydrogen in Ageing and Ageing-Related Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2249749. [PMID: 35340218 PMCID: PMC8956398 DOI: 10.1155/2022/2249749] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 02/10/2022] [Accepted: 03/03/2022] [Indexed: 12/17/2022]
Abstract
Ageing is a physiological process of progressive decline in the organism function over time. It affects every organ in the body and is a significant risk for chronic diseases. Molecular hydrogen has therapeutic and preventive effects on various organs. It has antioxidative properties as it directly neutralizes hydroxyl radicals and reduces peroxynitrite level. It also activates Nrf2 and HO-1, which regulate many antioxidant enzymes and proteasomes. Through its antioxidative effect, hydrogen maintains genomic stability, mitigates cellular senescence, and takes part in histone modification, telomere maintenance, and proteostasis. In addition, hydrogen may prevent inflammation and regulate the nutrient-sensing mTOR system, autophagy, apoptosis, and mitochondria, which are all factors related to ageing. Hydrogen can also be used for prevention and treatment of various ageing-related diseases, such as neurodegenerative disorders, cardiovascular disease, pulmonary disease, diabetes, and cancer. This paper reviews the basic research and recent application of hydrogen in order to support hydrogen use in medicine for ageing prevention and ageing-related disease therapy.
Collapse
|
11
|
Luo P, Ding Y, He Y, Chen D, He Q, Huang Z, Huang S, Lei W. Hydrogen-oxygen therapy alleviates clinical symptoms in twelve patients hospitalized with COVID-19: A retrospective study of medical records. Medicine (Baltimore) 2022; 101:e27759. [PMID: 35244034 PMCID: PMC8896485 DOI: 10.1097/md.0000000000027759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 10/28/2021] [Indexed: 01/04/2023] Open
Abstract
A global public health crisis caused by the 2019 novel coronavirus disease (COVID-19) leads to considerable morbidity and mortality, which bring great challenge to respiratory medicine. Hydrogen-oxygen therapy contributes to treat severe respiratory diseases and improve lung functions, yet there is no information to support the clinical use of this therapy in the COVID-19 pneumonia.A retrospective study of medical records was carried out in Shishou Hospital of Traditional Chinese Medicine in Hubei, China. COVID-19 patients (aged ≥ 30 years) admitted to the hospital from January 29 to March 20, 2020 were subjected to control group (n = 12) who received routine therapy and case group (n = 12) who received additional hydrogen-oxygen therapy. The clinical characteristics of COVID-19 patients were analyzed. The physiological and biochemical indexes, including immune inflammation indicators, electrolytes, myocardial enzyme profile, and functions of liver and kidney, were examined and investigated before and after hydrogen-oxygen therapy.The results showed significant decreases in the neutrophil percentage and the concentration and abnormal proportion of C-reactive protein in COVID-19 patients received additional hydrogen-oxygen therapy.This novel therapeutic may alleviate clinical symptoms of COVID-19 patients by suppressing inflammation responses.
Collapse
Affiliation(s)
- Peng Luo
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yuanfang Ding
- Cardiology Department, Shishou Hospital of Traditional Chinese Medcine, Jingzhou, Hubei, China
| | - Yuan He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Dafeng Chen
- Department of Precision Laboratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Qing He
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Zufeng Huang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Cardiology Department, Shishou Hospital of Traditional Chinese Medcine, Jingzhou, Hubei, China
| | - Shian Huang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Wei Lei
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
- Laboratory of Cardiovascular Diseases, Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Precision Laboratory Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
12
|
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
|
13
|
Tian Y, Zhang Y, Wang Y, Chen Y, Fan W, Zhou J, Qiao J, Wei Y. Hydrogen, a Novel Therapeutic Molecule, Regulates Oxidative Stress, Inflammation, and Apoptosis. Front Physiol 2022; 12:789507. [PMID: 34987419 PMCID: PMC8721893 DOI: 10.3389/fphys.2021.789507] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/22/2021] [Indexed: 12/21/2022] Open
Abstract
Molecular hydrogen (H2) is a colorless and odorless gas. Studies have shown that H2 inhalation has the therapeutic effects in many animal studies and clinical trials, and its application is recommended in the novel coronavirus pneumonia treatment guidelines in China recently. H2 has a relatively small molecular mass, which helps it quickly spread and penetrate cell membranes to exert a wide range of biological effects. It may play a role in the treatment and prevention of a variety of acute and chronic inflammatory diseases, such as acute pancreatitis, sepsis, respiratory disease, ischemia reperfusion injury diseases, autoimmunity diseases, etc.. H2 is primarily administered via inhalation, drinking H2-rich water, or injection of H2 saline. It may participate in the anti-inflammatory and antioxidant activity (mitochondrial energy metabolism), immune system regulation, and cell death (apoptosis, autophagy, and pyroptosis) through annihilating excess reactive oxygen species production and modulating nuclear transcription factor. However, the underlying mechanism of H2 has not yet been fully revealed. Owing to its safety and potential efficacy, H2 has a promising potential for clinical use against many diseases. This review will demonstrate the role of H2 in antioxidative, anti-inflammatory, and antiapoptotic effects and its underlying mechanism, particularly in coronavirus disease-2019 (COVID-19), providing strategies for the medical application of H2 for various diseases.
Collapse
Affiliation(s)
- Yan Tian
- Research Center for Translational Medicine, Tongji University Affiliated East Hospital, Shanghai, China
| | - Yafang Zhang
- Department of Pediatrics, Taian City Central Hospital, Taian, China
| | - Yu Wang
- Research Center for Translational Medicine, Tongji University Affiliated East Hospital, Shanghai, China.,Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Yunxi Chen
- Research Center for Translational Medicine, Tongji University Affiliated East Hospital, Shanghai, China
| | - Weiping Fan
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Jianjun Zhou
- Research Center for Translational Medicine, Tongji University Affiliated East Hospital, Shanghai, China
| | - Jing Qiao
- Department of Pediatrics, Tongji University Affiliated East Hospital, Shanghai, China
| | - Youzhen Wei
- Research Center for Translational Medicine, Tongji University Affiliated East Hospital, Shanghai, China
| |
Collapse
|
14
|
Du J, Li J, Li R, Yan X. High concentration of hydrogen ameliorates lipopolysaccharide-induced acute lung injury in a sirt1-dependent manner. Respir Physiol Neurobiol 2021; 296:103808. [PMID: 34757082 DOI: 10.1016/j.resp.2021.103808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/20/2021] [Accepted: 10/24/2021] [Indexed: 01/28/2023]
Abstract
The aim of this study was to investigate the efficacy and underlying mechanism of high concentration of hydrogen on lipopolysaccharide (LPS)-induced acute lung injury (ALI). We have established a corresponding mouse model and examined the function of hydrogen inhalation on lung pathology and pulmonary edema induced by LPS, as well as contents of IL-1β, TNF-α and IL-8. The pulmonary microvascular permeability and 66.7 % hydrogen on the expression of sirt1 and its downstream signaling molecules were tested. Results showed that 66.7 % hydrogen alleviated lung pathological changes and pulmonary edema caused by LPS, and reduced the degree of ALI by inhibiting pro-inflammatory cytokine release and oxidative stress response, thereby decreasing the expression of molecules related to intercellular adhesion. sirt1 contributed to the repair of LPS-induced ALI by hydrogen through the regulation of NF-κB and catalase expression. In conclusion, 66.7 % hydrogen protected against LPS-induced ALI by suppressing inflammatory response and oxidative stress mediated by NF-κB and catalase in a sirt1-dependent manner.
Collapse
Affiliation(s)
- Junfeng Du
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China; Department of Respiratory and Critical Care Medicine, Cangzhou Central Hospital, Cangzhou, Cangzhou 061001, China
| | - Jingwen Li
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China; Hebei Key Laboratory of Respiratory and Critical Diseases, Shijiazhuang 050000, China
| | - Rongqin Li
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Xixin Yan
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China; Hebei Key Laboratory of Respiratory and Critical Diseases, Shijiazhuang 050000, China; Hebei Provincial Institute of Respiratory Diseases, Shijiazhuang 050000, China.
| |
Collapse
|
15
|
Sun R, Zhao N, Wang Y, Su Y, Zhang J, Wang Y, Yu Y, Wang G, Wang Z, Xie K. High concentration of hydrogen gas alleviates Lipopolysaccharide-induced lung injury via activating Nrf2 signaling pathway in mice. Int Immunopharmacol 2021; 101:108198. [PMID: 34634688 DOI: 10.1016/j.intimp.2021.108198] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND AIMS The lung is the first organ to fail in sepsis. Our previous studies have proven that 2% molecular hydrogen (H2) inhalation remain a protective effect on a septic animal model via its anti-inflammatory and anti-apoptosis properties. This current research aims to observe the therapeutic effect of high concentration hydrogen (67%, HCH) on lipopolysaccharide (LPS) induced acute lung injury (ALI), and further investgate the role of Nrf2 signaling pathway. METHODS ALI model was induced by LPS areosol inhalation. HCH were treated for 1 h at 1 and 6 h after modelling. Lung tissues and bronchoalveolar lavage fluid (BALF) were collected 4 and 24 h after the exposure of LPS. The histological scores, wet/dry weight ratios, myeloperoxidase (MPO) activity, protein content and cytokine levels in BALF, apoptosis condition of lung cells, expression of Nrf2 and NF-κB were assessed in both wild type and Nrf2-knockout mice. RESULTS HCH Inhalation significantly alleviated LPS-induced pathological alterations of lung, and reduced the protein concentration, the wet/dry weight ratio, and the MPO activity of lung tissue. HCH Inhalation improved LPS-induced increasement in caspase-3 activity and the number of TUNEL-positive cells. HCH inhalation attenuated the LPS induced increased total cell content and polymorphonuclear granulocyte content, and pro-inflammatory cytokines, Nrf2 and NF-κB expression. HCH could not produce protective effct in Nrf2-knockout mice. CONCLUSION HCH can effectively alleviate LPS-induced ALI, which may be related to activation of Nrf2 signaling pathway and inhibition of inflammatory response and cell apoptosis mediated by NF-κB.
Collapse
Affiliation(s)
- Ruiqiang Sun
- Department of Anesthesiology, Tianjin Eye Hospital, Tianjin 300020, China; Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Nan Zhao
- Department of Anesthesiology, Tianjin Chest Hospital, Tianjin 300222, China
| | - Yuzun Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Yanchao Su
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jiayan Zhang
- Department of Critical Care Medicine, 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
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhen Wang
- 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; Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China; Translational Research Institute of Intensive Care Medicine, College of Anesthesiology, Weifang Medical University, Weifang 261053, Shandong Province, China.
| |
Collapse
|
16
|
Zheng P, Kang J, Xing E, Zheng B, Wang X, Zhou H. Lung Inflation With Hydrogen During the Cold Ischemia Phase Alleviates Lung Ischemia-Reperfusion Injury by Inhibiting Pyroptosis in Rats. Front Physiol 2021; 12:699344. [PMID: 34408660 PMCID: PMC8365359 DOI: 10.3389/fphys.2021.699344] [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: 04/23/2021] [Accepted: 07/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Lung inflation with hydrogen is an effective method to protect donor lungs from lung ischemia-reperfusion injury (IRI). This study aimed to examine the effect of lung inflation with 3% hydrogen during the cold ischemia phase on pyroptosis in lung grafts of rats. Methods: Adult male Wistar rats were randomly divided into the sham group, the control group, the oxygen (O2) group, and the hydrogen (H2) group. The sham group underwent thoracotomy but no lung transplantation. In the control group, the donor lungs were deflated for 2 h. In the O2 and H2 groups, the donor lungs were inflated with 40% O2 + 60% N2 and 3% H2 + 40% O2 + 57% N2, respectively, at 10 ml/kg, and the gas was replaced every 20 min during the cold ischemia phase for 2 h. Two hours after orthotopic lung transplantation, the recipients were euthanized. Results: Compared with the control group, the O2 and H2 groups improved oxygenation indices, decreases the inflammatory response and oxidative stress, reduced lung injury, and improved pressure-volume (P-V) curves. H2 had a better protective effect than O2. Furthermore, the levels of the pyroptosis-related proteins selective nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3), cysteinyl aspartate specific proteinase (caspase)-1 p20, and the N-terminal of gasdermin D (GSDMD-N) were decreased in the H2 group. Conclusion: Lung inflation with 3% hydrogen during the cold ischemia phase inhibited the inflammatory response, oxidative stress, and pyroptosis and improved the function of the graft. Inhibiting reactive oxygen species (ROS) production may be the main mechanism of the antipyroptotic effect of hydrogen.
Collapse
Affiliation(s)
- Panpan Zheng
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiyu Kang
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Entong Xing
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Bin Zheng
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Xueyao Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Huacheng Zhou
- Department of Anesthesiology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Su JC, Zhang Y, Cheng C, Zhu YN, Ye YM, Sun YK, Xiang SY, Wang Y, Liu ZB, Zhang XF. Hydrogen regulates the M1/M2 polarization of alveolar macrophages in a rat model of chronic obstructive pulmonary disease. Exp Lung Res 2021; 47:301-310. [PMID: 34282696 DOI: 10.1080/01902148.2021.1919788] [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: 10/20/2022]
Abstract
OBJECTIVE Chronic obstructive pulmonary disease (COPD) is a respiratory disease with high morbidity and mortality worldwide, so far there is no ideal treatment method. Previous studies have shown that hydrogen (H2) is involved in the treatment of COPD as an antioxidant. In this study, the effect of H2 on M1/M2 polarization of alveolar macrophages in COPD rats was observed, and its anti-inflammatory mechanism was further elucidated. Methods: Twenty-four Sprague-Dawley rats were randomly divided into three groups including the control, COPD and H2 group. A rat model of COPD was established by cigarette exposure combined with lipopolysaccharide (LPS) induction. H2 therapy was administered 2 hours per day for 14 days. Lung function and pathology were assessed. The levels of interleukin (IL)-6, tumor necrosis factor (TNF)-α, transforming growth factor (TGF)-β1 and IL-10 in bronchoalveolar lavage fluid (BALF) and lung tissue were measured by enzyme-linked immunosorbent assay. The mRNA, protein expression and immunoreactivity of inducible nitric oxide synthase (iNOS) and arginase (Arg)-1 in lung were observed by quantitative real-time PCR, western blot and immunohistochemistry. Results: Compared with the control rats, there were a significant decline in lung function, a marked inflammatory infiltration and pulmonary parenchymal remodeling and the increases of IL-6, TNF-α and TGF-β1 levels in BALF and lung tissue, but a lower expression of IL-10 in COPD rats. The iNOS mRNA and protein expression, as well as its optical density (OD), were increased significantly in lung tissue, while those of Arg-1 decreased significantly. H2 treatment improved the lung function and the parenchymal inflammation, reversed the increased levels of IL-6, TNF-α and TGF-β1, and the lower IL-10. Meanwhile, H2 also down-regulated the expression of iNOS, but up-regulated expression of Arg-1 in lung tissue. Conclusion: H2 reduces inflammation in the lung of COPD, which may be related to its inhibition of M1 type polarization and activation of M2 type polarization of alveolar macrophage.
Collapse
Affiliation(s)
- Jing-Chao Su
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yi Zhang
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, China.,College of Acupuncture and Tuina, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Chen Cheng
- Graduate School, Anhui University of Chinese Medicine, Hefei, Anhui, China.,College of Acupuncture and Tuina, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yi-Nan Zhu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yu-Meng Ye
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yong-Kang Sun
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Shui-Ying Xiang
- College of Acupuncture and Tuina, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yuan Wang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Zi-Bing Liu
- College of Acupuncture and Tuina, Anhui University of Chinese Medicine, Hefei, Anhui, China.,Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xin-Fang Zhang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.,Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
18
|
Chen JB, Kong XF, Mu F, Lu TY, Lu YY, Xu KC. Hydrogen therapy can be used to control tumor progression and alleviate the adverse events of medications in patients with advanced non-small cell lung cancer. Med Gas Res 2021; 10:75-80. [PMID: 32541132 PMCID: PMC7885710 DOI: 10.4103/2045-9912.285560] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy, targeted therapy, and immunotherapy are used against advanced non-small cell lung cancer. A clinically efficacious method for relieving the adverse events associated of such therapies is lacking. Fifty-eight adult patients were enrolled in our trial to relieve pulmonary symptoms or the adverse events of drugs. Twenty patients who refused drug treatment were assigned equally and randomly to a hydrogen (H2)-only group and a control group. According to the results of tumor-gene mutations and drug-sensitivity tests, 10, 18, and 10 patients were enrolled into chemotherapy, targeted therapy, and immunotherapy groups in which these therapies were combined with H2-therapy, respectively. Patients underwent H2 inhalation for 4–5 hours per day for 5 months or stopped when cancer recurrence. Before study initiation, the demographics (except for tumor-mutation genes) and pulmonary symptoms (except for moderate cough) of the five groups showed no significant difference. During the first 5 months of treatment, the prevalence of symptoms of the control group increased gradually, whereas that of the four treatment groups decreased gradually. After 16 months of follow-up, progression-free survival of the control group was lower than that of the H2-only group, and significantly lower than that of H2 + chemotherapy, H2 + targeted therapy, and H2 + immunotherapy groups. In the combined-therapy groups, most drug-associated adverse events decreased gradually or even disappeared. H2 inhalation was first discovered in the clinic that can be used to control tumor progression and alleviate the adverse events of medications for patients with advanced non-small cell lung cancer. This study was approved by the Ethics Committee of Fuda Cancer Hospital of Jinan University on December 7, 2018 (approval No. Fuda20181207), and was registered at ClinicalTrials.gov (Identifier: NCT03818347) on January 28, 2019.
Collapse
Affiliation(s)
- Ji-Bing Chen
- Fuda Cancer Hospital of Jinan University, Guangzhou; Fuda Cancer Institute, Guangzhou, Guangdong Province, China
| | - Xiao-Feng Kong
- Fuda Cancer Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Feng Mu
- Fuda Cancer Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Tian-Yu Lu
- Fuda Cancer Hospital of Jinan University, Guangzhou; Fuda Cancer Institute, Guangzhou, Guangdong Province, China
| | - You-Yong Lu
- Central Lab, Beijing Cancer Hospital, Beijing, China
| | - Ke-Cheng Xu
- Fuda Cancer Hospital of Jinan University, Guangzhou; Fuda Cancer Institute, Guangzhou, Guangdong Province, China
| |
Collapse
|
19
|
Qin S. Role of Hydrogen in Atherosclerotic Disease: From Bench to Bedside. Curr Pharm Des 2021; 27:713-722. [PMID: 33234094 DOI: 10.2174/1381612826666201124112152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/22/2020] [Indexed: 12/08/2022]
Abstract
Atherosclerotic cardiovascular and cerebrovascular diseases are among the leading causes of morbidity and mortality worldwide. Given our recent understanding of its role as a small-molecule antioxidant and anti- inflammatory agent, hydrogen may play an important role in preventing and treating atherosclerotic cardiovascular and cerebrovascular disease. In the past decade, more than 50 publications in the English language literature considered the role of hydrogen as an anti-atherosclerotic agent. In this review, we summarized the pathophysiological characteristics and risk factors associated with atherosclerosis (AS) and the laboratory research data that focuses on hydrogen to prevent and treat this condition, including the responses observed in both animal models and human studies. We will also consider the molecular mechanisms underlying the efficacy of hydrogen molecules with respect to atherosclerotic disease. Future studies might include clinical trials with larger sample populations as well as experiments designed to explore the molecular mechanisms associated with hydrogen treatment in greater depth.
Collapse
Affiliation(s)
- Shucun Qin
- The Institute of Atherosclerosis and Taishan Institute for Hydrogen Biomedicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian 271000, China
| |
Collapse
|
20
|
Zhang L, Yu H, Tu Q, He Q, Huang N. New Approaches for Hydrogen Therapy of Various Diseases. Curr Pharm Des 2021; 27:636-649. [PMID: 33308113 DOI: 10.2174/1381612826666201211114141] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 10/02/2020] [Indexed: 11/22/2022]
Abstract
Hydrogen therapy has recently received increasing attention as an emerging and promising therapeutic technology due to its selective antioxidant property and cell energy regulatory capability in vivo. To solve the low solubility issue of hydrogen, a variety of nanomaterials and devices for hydrogen supply have recently been developed, aiming to increase the concentration of hydrogen in the specific disease site and realize controlled hydrogen release and combined treatment. In this review, we mainly focus on the latest advances in using hydrogen-generating devices and nanomaterials for hydrogen therapy. These developments include sustained release of H2, controlled release of H2, versatile modalities of synergistic therapy, etc. Also, bio-safety issues and challenges are discussed to further promote the clinical applications of hydrogen therapy and the development of hydrogen medicine.
Collapse
Affiliation(s)
- Lei Zhang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Han Yu
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Qiufen Tu
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Qianjun He
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Nan Huang
- School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
21
|
Lin HY, Lai PC, Chen WL. A narrative review of hydrogen-oxygen mixture for medical purpose and the inhaler thereof. Med Gas Res 2021; 10:193-200. [PMID: 33380588 PMCID: PMC8092144 DOI: 10.4103/2045-9912.295226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Recent development regarding mixture of H2 (concentration of ~66%) with O2 (concentration of ~34%) for medical purpose, such as treatment of coronavirus disease-19 (COVID-19) patients, is introduced. Furthermore, the design principles of a hydrogen inhaler which generates mixture of hydrogen (~66%) with oxygen (~34%) for medical purpose are proposed. With the installation of the liquid blocking module and flame arresters, the air pathway of the hydrogen inhaler is divided by multiple isolation zones to prevent any unexpected explosion propagating from one zone to the other. An integrated filtering/cycling module is utilized to purify the impurity, and cool down the temperature of the electrolytic module to reduce the risk of the explosion. Moreover, a nebulizer is provided to selectively atomize the water into vapor which is then mixed with the filtered hydrogen-oxygen mix gas, such that the static electricity of a substance hardly occurs to reduce the risk of the explosion. Furthermore, hydrogen concentration detector is installed to reduce the risk of hydrogen leakage. Result shows that the hydrogen inhaler implementing the aforesaid design rules could effectively inhibit the explosion, even ignition at the outset of the hydrogen inhaler which outputs hydrogen-oxygen gas (approximately 66% hydrogen: 34% oxygen).
Collapse
|
22
|
Wang ST, Bao C, He Y, Tian X, Yang Y, Zhang T, Xu KF. Hydrogen gas (XEN) inhalation ameliorates airway inflammation in asthma and COPD patients. QJM 2020; 113:870-875. [PMID: 32407476 PMCID: PMC7785302 DOI: 10.1093/qjmed/hcaa164] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/01/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Hydrogen was proven to have anti-oxidative and anti-inflammation effects to various diseases. AIM We wish to investigate the acute effects of inhaled hydrogen on airway inflammation in patients with asthma and chronic obstructive pulmonary disease (COPD). DESIGN Prospective study. METHODS In total, 2.4% hydrogen containing steam mixed gas (XEN) was inhaled once for 45 min in 10 patients with asthma and 10 patients with COPD. The levels of granulocyte-macrophage colony stimulating factor, interferon-γ, interleukin-1β (IL-1β), IL-2, IL-4, IL-6 and so on in peripheral blood and exhaled breath condensate (EBC) before and after 'XEN' inhalation were measured. RESULTS 45 minutes 'XEN' inhalation once decreased monocyte chemotactic protein 1 level in both COPD (564.70-451.51 pg/mL, P = 0.019) and asthma (386.39-332.76 pg/mL, P = 0.033) group, while decreased IL-8 level only in asthma group (5.25-4.49 pg/mL, P = 0.023). The level of EBC soluble cluster of differentiation-40 ligand in COPD group increased after inhalation (1.07-1.16 pg/mL, P = 0.031), while IL-4 and IL-6 levels in EBC were significantly lower after inhalation in the COPD (0.80-0.64 pg/mL, P = 0.025) and asthma (0.06-0.05 pg/mL, P = 0.007) group, respectively. CONCLUSIONS A single inhalation of hydrogen for 45 min attenuated inflammatory status in airways in patients with asthma and COPD.
Collapse
Affiliation(s)
- S -T Wang
- From the Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - C Bao
- From the Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Y He
- From the Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - X Tian
- From the Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Y Yang
- From the Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - T Zhang
- From the Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - K -F Xu
- From the Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
23
|
Qi C, Zhou J, Wang Z, Fang X, Li D, Jin Y, Song J. Cigarette smoke extract combined with lipopolysaccharide reduces OCTN1/2 expression in human alveolar epithelial cells in vitro and rat lung in vivo under inflammatory conditions. Int Immunopharmacol 2020; 87:106812. [PMID: 32707498 DOI: 10.1016/j.intimp.2020.106812] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/25/2020] [Accepted: 07/13/2020] [Indexed: 12/21/2022]
Abstract
Organic cation transporter 1/2 (OCTN1/2) play important roles in the transport of drugs related to pulmonary inflammatory diseases. Nevertheless, the involvement of inflammation induced by cigarette smoke extract (CSE) combined with lipopolysaccharide (LPS) in the regulation of OCTN1/2 is not fully understood. In this study, CSE combined with LPS was used to establish inflammation models in vitro and in vivo. Our study found that the expression of OCTN1/2 was downregulated in rat lung in vivo and in a human alveolar cell line in vitro after treatment with CSE and LPS compared with the control group, while the expression of inflammatory factors was upregulated. After treatment with ipratropium bromide (IB) or dexamethasone (DEX), the expression of OCTN1/2 was upregulated compared with that in the CSE-LPS model group, while the expression of inflammatory factors was significantly downregulated. After administration of the NF-κB inhibitor PDTC on the basis of the inflammatory status, the expression of OCTN1/2 was upregulated in the treated group compared with the CSE-LPS model group, while the expression of phospho-p65, phospho-IκBα and inflammatory factors was significantly downregulated. We further added the NF-κB agonist HSP70 and found a result that the exact opposite of that observed with PDTC. Our findings show that CSE combined with LPS can downregulate the expression of OCTN1/2 under inflammatory conditions, and that the downregulation of OCTN1/2 expression may partially occur via the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chuanzong Qi
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Jian Zhou
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Zihao Wang
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Xin Fang
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Dalang Li
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Yong Jin
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China
| | - Jue Song
- Institute for Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; Institute for the Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, China.
| |
Collapse
|
24
|
Feng S, Duan E, Shi X, Zhang H, Li H, Zhao Y, Chao L, Zhong X, Zhang W, Li R, Yan X. Hydrogen ameliorates lung injury in a rat model of subacute exposure to concentrated ambient PM2.5 via Aryl hydrocarbon receptor. Int Immunopharmacol 2019; 77:105939. [DOI: 10.1016/j.intimp.2019.105939] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 08/22/2019] [Accepted: 09/26/2019] [Indexed: 01/16/2023]
|
25
|
Huang L, Applegate RL, Applegate PM, Boling W, Zhang JH. Inhalation of high concentration hydrogen gas improves short-term outcomes in a rat model of asphyxia induced-cardiac arrest. Med Gas Res 2018; 8:73-78. [PMID: 30319760 PMCID: PMC6178639 DOI: 10.4103/2045-9912.241063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 06/14/2018] [Indexed: 12/18/2022] Open
Abstract
Cardiogenic global brain hypoxia-ischemia is a devastating medical problem that is associated with unfavorable neurologic outcomes. Low dose hydrogen gas (up to 2.9%) has been shown to be neuroprotective in a variety of brain diseases. In the present study, we investigated the protective effect of water by electrolysis-derived high concentration hydrogen gas (60%) in a rat model of asphyxia induced-cardiac arrest and global brain hypoxia-ischemia. High concentration hydrogen gas was either administered starting 1 hour prior to cardiac arrest for 1 hour and starting 1 hour post-resuscitation for 1 hour (pre- & post-treatment) or starting 1 hour post-resuscitation for 2 hours (post-treatment). In animals subjected to 9 minutes of asphyxia, both therapeutic regimens tended to reduce the incidence of seizures and neurological deficits within 3 days post-resuscitation. In rats subjected to 11 minutes of asphyxia, significantly worse neurological deficits were observed compared to 9 minutes asphyxia, and pre- & post-treatment had a tendency to improve the success rate of resuscitation and to reduce the seizure incidence within 3 days post-resuscitation. Findings of this preclinical study suggest that water electrolysis-derived 60% hydrogen gas may improve short-term outcomes in cardiogenic global brain hypoxia-ischemia.
Collapse
Affiliation(s)
- Lei Huang
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Basic Science, Division of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Richard L Applegate
- Department of Anesthesiology and Pain Medicine, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Patricia M Applegate
- Department of Cardiology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Warren Boling
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Basic Science, Division of Physiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
26
|
Lu W, Li D, Hu J, Mei H, Shu J, Long Z, Yuan L, Li D, Guan R, Li Y, Xu J, Wang T, Yao H, Zhong N, Zheng Z. Hydrogen gas inhalation protects against cigarette smoke-induced COPD development in mice. J Thorac Dis 2018; 10:3232-3243. [PMID: 30069319 DOI: 10.21037/jtd.2018.05.93] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Chronic obstructive pulmonary disease (COPD) is a chronic lung disease with limited treatment options. Hydrogen (H2) has been shown to be anti-oxidative and anti-inflammatory. This study aimed to evaluate the beneficial effects of H2 inhalation on COPD development in mice. Methods A COPD mouse model was established in male C57BL mice by cigarette smoke (CS) exposure. The H2 intervention was administered by atomisation inhalation. Lung functions were assessed by using Buxco lung function measurement system. The inflammatory cells were counted and the levels of IL-6 and KC in BALF were assayed with ELISA. The lung tissue was subjected to H&E or PAS or Masson's trichrome stain. Furthermore, 16HBE cells were used to evaluate the effects of H2 on signaling change caused by hydrogen peroxide (H2O2). H2O2 was used to treat 16HBE cells with or without H2 pretreatment. The IL-6 and IL-8 levels in cell culture medium were measured. The levels of phosphorylated ERK1/2 and nucleic NF-κB in lungs and 16HBE cells were determined. Results H2 ameliorated CS-induced lung function decline, emphysema, inflammatory cell infiltration, small-airway remodelling, goblet-cell hyperplasia in tracheal epithelium and activated ERK1/2 and NF-κB in mouse lung. In 16HBE airway cells, H2O2 increased IL-6 and IL-8 secretion in conjunction with ERK1/2 and NF-κB activation. These changes were reduced by H2 treatment. Conclusions These findings demonstrated that H2 inhalation could inhibit CS-induced COPD development in mice, which is associated with reduced ERK1/2 and NF-κB-dependent inflammatory responses.
Collapse
Affiliation(s)
- Wenju Lu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China.,Department of Laboratory Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China.,Sino-French Hoffmann Immunology Institute, Guangzhou Medical University, Guangzhou 510000, China
| | - Defu Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Jieying Hu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Huijun Mei
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Jiaze Shu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Zhen Long
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Liang Yuan
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Difei Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Ruijuan Guan
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Yuanyuan Li
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Jingyi Xu
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Tao Wang
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Hongwei Yao
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| | - Zeguang Zheng
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China.,Department of Laboratory Medicine, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou 510000, China
| |
Collapse
|
27
|
Weidner J, Jarenbäck L, Åberg I, Westergren‐Thorsson G, Ankerst J, Bjermer L, Tufvesson E. Endoplasmic reticulum, Golgi, and lysosomes are disorganized in lung fibroblasts from chronic obstructive pulmonary disease patients. Physiol Rep 2018; 6:e13584. [PMID: 29484832 PMCID: PMC5827558 DOI: 10.14814/phy2.13584] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/18/2017] [Accepted: 12/26/2017] [Indexed: 12/23/2022] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is often caused by smoking and other stressors. This causes oxidative stress, which induces numerous changes on both the transcriptome and proteome of the cell. We aimed to examine if the endomembrane pathway, including the endoplasmic reticulum (ER), Golgi, and lysosomes, was disrupted in fibroblasts from COPD patients as opposed to healthy ever-smokers or never-smokers, and if the response to stress differed. Different cellular compartments involved in the endomembrane pathway, as well as mRNA expression and apoptosis, were examined before and after the addition of stress in lung fibroblasts from never-smokers, ever-smokers, and patients with COPD. We found that the ER, Golgi, and lysosomes were disorganized in fibroblasts from COPD patients under baseline conditions. After a time course with ER stress inducing chemicals, changes to the phenotypes of cellular compartments in COPD patient fibroblasts were observed, and the expression of the ER stress-induced gene ERP72 was upregulated more in the COPD patient's cells compared to ever-smokers or never-smokers. Lastly, a tendency of increased active Caspase-3 was observed in COPD fibroblasts. Our results show that COPD patients have phenotypic changes in the lung fibroblasts endomembrane pathway, and respond differently to stress. Furthermore, these fibroblasts were cultured for several weeks outside the body, but they were not able to regain proper ER structure, indicating that the internal changes to the endomembrane system are permanent in smokers. This vulnerability to cellular stress might be a cause as to why some smokers develop COPD.
Collapse
Affiliation(s)
- Julie Weidner
- Department of Clinical Sciences Lund, Respiratory Medicine and AllergologyLund UniversityLundSweden
| | - Linnea Jarenbäck
- Department of Clinical Sciences Lund, Respiratory Medicine and AllergologyLund UniversityLundSweden
| | - Ida Åberg
- Department of Clinical Sciences Lund, Respiratory Medicine and AllergologyLund UniversityLundSweden
| | | | - Jaro Ankerst
- Department of Clinical Sciences Lund, Respiratory Medicine and AllergologyLund UniversityLundSweden
| | - Leif Bjermer
- Department of Clinical Sciences Lund, Respiratory Medicine and AllergologyLund UniversityLundSweden
| | - Ellen Tufvesson
- Department of Clinical Sciences Lund, Respiratory Medicine and AllergologyLund UniversityLundSweden
| |
Collapse
|
28
|
Hydrogen-rich pure water prevents cigarette smoke-induced pulmonary emphysema in SMP30 knockout mice. Biochem Biophys Res Commun 2017; 492:74-81. [PMID: 28807355 DOI: 10.1016/j.bbrc.2017.08.035] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 08/10/2017] [Indexed: 10/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is predominantly a cigarette smoke (CS)-triggered disease with features of chronic systemic inflammation. Oxidants derived from CS can induce DNA damage and stress-induced premature cellular senescence in the respiratory system, which play significant roles in COPD. Therefore, antioxidants should provide benefits for the treatment of COPD; however, their therapeutic potential remains limited owing to the complexity of this disease. Recently, molecular hydrogen (H2) has been reported as a preventive and therapeutic antioxidant. Molecular H2 can selectively reduce hydroxyl radical accumulation with no known side effects, showing potential applications in managing oxidative stress, inflammation, apoptosis, and lipid metabolism. However, there have been no reports on the efficacy of molecular H2 in COPD patients. In the present study, we used a mouse model of COPD to investigate whether CS-induced histological damage in the lungs could be attenuated by administration of molecular H2. We administered H2-rich pure water to senescence marker protein 30 knockout (SMP30-KO) mice exposed to CS for 8 weeks. Administration of H2-rich water attenuated the CS-induced lung damage in the SMP30-KO mice and reduced the mean linear intercept and destructive index of the lungs. Moreover, H2-rich water significantly restored the static lung compliance in the CS-exposed mice compared with that in the CS-exposed H2-untreated mice. Moreover, treatment with H2-rich water decreased the levels of oxidative DNA damage markers such as phosphorylated histone H2AX and 8-hydroxy-2'-deoxyguanosine, and senescence markers such as cyclin-dependent kinase inhibitor 2A, cyclin-dependent kinase inhibitor 1, and β-galactosidase in the CS-exposed mice. These results demonstrated that H2-rich pure water attenuated CS-induced emphysema in SMP30-KO mice by reducing CS-induced oxidative DNA damage and premature cell senescence in the lungs. Our study suggests that administration of molecular H2 may be a novel preventive and therapeutic strategy for COPD.
Collapse
|