1
|
Liao S, Chen D, Long H, Jiang S, Fan J, Li S, Qi Y, Xue L, Ding Y, Chen Y. Hydrogen sulfide attenuates oxidative stress-induced cellular senescence via the Sirt3/SOD2 signaling pathway in chronic obstructive pulmonary disease. Chin Med J (Engl) 2025:00029330-990000000-01470. [PMID: 40082252 DOI: 10.1097/cm9.0000000000003452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Senescence significantly participates in shaping the pathobiological process underlying chronic obstructive pulmonary disease (COPD). Currently, the mechanisms underlying the anti-aging effects within COPD of hydrogen sulfide (H2S) are not fully illustrated. METHODS Immunohistochemistry (IHC) staining was performed on human lung tissue to detect the expression levels of sirtuin 3 (Sirt3), cyclin-dependent kinase 4 inhibitor (P16), and cystathionin gamma lyase (CTH). An animal model including wild-type (WT) and Sirt3 knockout (KO) mice was established by exposing them to cigarette smoking (CS) for 24 weeks, with or without intraperitoneal injection of sodium hydrosulfide (NaHS, 50 µmol∙L-1∙kg-1) 30 min prior to CS exposure. Lung function was assessed. The expression levels of P16, cyclin-dependent kinase inhibitor 1 (P21), Sirt3, manganese superoxide dismutase (SOD2), manganese acetylated superoxide dismutase (ac-SOD2), interleukin-6 (IL-6), IL-8, malondialdehyde (MDA), and glutathione (GSH), as well as the activity of SOD2 and Sirt3, were evaluated. Human bronchial epithelial BEAS-2B cells were subjected to diverse cigarette smoking extract (CSE) concentrations for 48 h with or without sodium hydrosulfide (NaHS). Subsequently, the levels of total intracellular reactive oxygen species (T-ROS), mitochondrial reactive oxygen species (mitoROS), mitochondrial membrane potential (MMP), senescence-associated β-galactosidase (SA-β-gal) staining positive cells, and related marker proteins and cytokines were assessed. Furthermore, the Sirt3-specific inhibitor 3-TYP and small interfering RNAs (siRNAs) of Sirt3 were used to examine the mechanisms whereby H2S inhibits oxidative stress and senescence in COPD. RESULTS IHC showed a significant reduction of CTH and Sirt3 protein levels in the lung tissue of COPD with smoking patients and smokers without COPD compared to non-smokers. Furthermore, the expression of the aging marker protein P16 was notably elevated in the COPD with smoking group compared to the smokers without COPD and non-smoker groups. Furthermore, our results demonstrated that exposure to CS resulted in imbalanced oxidative and cellular senescence, including elevated mitoROS, T-ROS, MDA, and ac-SOD2, along with increased proportions of SA-β-gal staining positive cells and the increased expression levels of IL-6, IL-8, P21, and P16, as well as decreased GSH levels, SOD2 and Sirt3 activities, and Sirt3 expression, which ultimately contribute to emphysema development and impaired lung function. However, pretreatment with NaHS effectively reversed these detrimental effects. Nevertheless, the protective effect of NaHS was alleviated in Sirt3 KO mice and in cellular models treated with Sirt3 siRNA and 3-TYP. CONCLUSION Our study indicates that H2S inhibits oxidative stress and cellular senescence by modulating the Sirt3/SOD2 signaling pathway, therefore attenuating the emphysema and impaired lung function induced by CS.
Collapse
Affiliation(s)
- Sha Liao
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Dian Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Huanyu Long
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Simin Jiang
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Jing Fan
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Shurun Li
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Yongfen Qi
- Department of Pathogenic Biology, Peking University School of Basic Medicine, Beijing 100191, China
| | - Lixiang Xue
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Yanling Ding
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
| | - Yahong Chen
- Department of Pulmonary and Critical Care Medicine, Peking University Third Hospital, Beijing 100191, China
- Research Center for Chronic Airway Diseases, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
2
|
Kavianinia M, Kalantar H, Salehcheh M, Khorsandi L, Shariati S, Mohtadi S, Khodayar MJ. Dimethyl fumarate effects on paraquat-induced hepatotoxicity in mice via anti-oxidative, anti-inflammatory, and anti-apoptotic activities. Sci Rep 2025; 15:3897. [PMID: 39890857 PMCID: PMC11785811 DOI: 10.1038/s41598-025-88461-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: 09/17/2024] [Accepted: 01/28/2025] [Indexed: 02/03/2025] Open
Abstract
Paraquat (PQ) toxicity is a common problem in the world, associated with oxidative stress, inflammation, and apoptosis. Therefore, the use of agents that reduce these disorders can be effective in the treatment of PQ toxicity. The protective effects of dimethyl fumarate (DMF) on liver disorders have been suggested in many reports. In this study, mice were divided into 6 groups; control, PQ (30 mg/kg, i.p., at day 4), DMF (100 mg/kg, p.o.), and PQ groups pretreated by DMF in three doses 10, 30, and 100 mg/kg, respectively. DMF was administered for 7 days to counteract PQ-induced liver toxicity. On the 8th day, mice were euthanized with ketamine/xylazine, and serum factors, oxidative stress markers, apoptosis index, and inflammatory markers were measured. PQ significantly increased the activity level of serum enzymes, thiobarbituric acid reactive substances, apoptotic factor (Bax/Bcl-2 ratio), inflammatory factors (NF-κB protein expression, tumor necrosis factor-α, interleukin-1β), nitric oxide, and Nrf-2 protein expression. Furthermore, PQ decreased hepatic total thiol and activity levels of catalase, superoxide dismutase, and glutathione peroxidase. However, DMF reduced the harmful effects caused by the imbalance in the oxidant and antioxidant system and histopathological damage in PQ-poisoned mice and improved the damage caused by inflammation and apoptosis.
Collapse
Affiliation(s)
- Maryam Kavianinia
- Medicinal Plant Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadi Kalantar
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Maryam Salehcheh
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeedeh Shariati
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shokooh Mohtadi
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Javad Khodayar
- Medicinal Plant Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Department of Toxicology, Faculty of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
3
|
Hammami H, Mozafarjalali M, Hajiani M. Plant extracts as an eco-friendly approach to remove paraquat from aqueous solution. INTERNATIONAL JOURNAL OF PHYTOREMEDIATION 2024; 26:1049-1063. [PMID: 38062787 DOI: 10.1080/15226514.2023.2288903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Nowadays, water pollution by herbicides is known as a global concern. Paraquat (PQ) (1-1-methyl-4,4-bi-pyridinium-dichloride) is a chip with high performance, which is being widely used herbicide to remove weeds from agricultural and natural ecosystems. PQ can contaminate water sources due to its high solubility in water. Human death by poisoning effects of PQ has been reported in several countries. Therefore, the side effects of PQ are a global challenge. This study aimed to investigate the bioremediation of PQ by plant extracts, as a low-cost, nontoxic, and natural absorbent to remove PQ from aqueous solutions in different conditions. In this regard, the extracts of common purslane (portulaca oleracea), florist kalanchoe (kalanchoe blossfeldiana), and jade plant (crassula portulaca) were used as adsorbents. For this purpose, the effect of various parameters such as contact time, initial concentration of PQ solution, temperature, pH, and amount of extract was investigated. The results of present study showed that P. oleracea extract and C. portulaca extracts have higher adsorption efficiency than k. blossfeldiana extract. The highest PQ removal was obtained by P. oleracea extract (79.04%) and C. portulaca extract (78.72%) at pH = 11, the adsorbent content of 0.2 mg L-1, and the lowest absorption of PQ (50.6%) was obtained by K. blossfeldiana extract. The highest PQ removal by plant extract was observed at 30 min for P. oleracea and C. portulaca, and at 15 min for k. blossfeldiana extract. Moreover, surface absorption capacity increased with increasing plant extract concentration, decreasing PQ concentration and decreased with increasing temperature. Finally, it can be concluded that plant extract can help to remove PQ from the aqueous solution.
Collapse
Affiliation(s)
- Hossein Hammami
- Department of Plant Production and Genetic Engineering, Faculty of Agriculture, University of Birjand, Birjand, Iran
- Member of the Plant and Environmental Stresses Research Group, University of Birjand, Birjand, Iran
- Member of the Unconventional Water Research Group, University of Birjand, Birjand, Iran
| | - Maryam Mozafarjalali
- Department of Environmental Engineering, Faculty of Natural Resources and Environment, University of Birjand, Birjand, Iran
| | - Mahmood Hajiani
- Department of Environmental Engineering, Faculty of Natural Resources and Environment, University of Birjand, Birjand, Iran
| |
Collapse
|
4
|
Zhang H, Chen N, Ding C, Zhang H, Liu D, Liu S. Ferroptosis and EMT resistance in cancer: a comprehensive review of the interplay. Front Oncol 2024; 14:1344290. [PMID: 38469234 PMCID: PMC10926930 DOI: 10.3389/fonc.2024.1344290] [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: 11/25/2023] [Accepted: 01/30/2024] [Indexed: 03/13/2024] Open
Abstract
Ferroptosis differs from traditional cell death mechanisms like apoptosis, necrosis, and autophagy, primarily due to its reliance on iron metabolism and the loss of glutathione peroxidase activity, leading to lipid peroxidation and cell death. The dysregulation of iron metabolism is a hallmark of various cancers, contributing to tumor progression, metastasis, and notably, drug resistance. The acquisition of mesenchymal characteristics by epithelial cells is known as Epithelial-Mesenchymal Transition (EMT), a biological process intricately linked to cancer development, promoting traits such as invasiveness, metastasis, and resistance to therapeutic interventions. EMT plays a pivotal role in cancer progression and contributes significantly to the complex dynamics of carcinogenesis. Research findings indicate that mesenchymal cancer cells exhibit greater susceptibility to ferroptosis compared to their epithelial counterparts. The induction of ferroptosis becomes more effective in eliminating drug-resistant cancer cells during the process of EMT. The interplay between ferroptosis and EMT, a process where epithelial cells transform into mobile mesenchymal cells, is crucial in understanding cancer progression. EMT is associated with increased cancer metastasis and drug resistance. The review delves into how ferroptosis and EMT influence each other, highlighting the role of key proteins like GPX4, which protects against lipid peroxidation, and its inhibition can induce ferroptosis. Conversely, increased GPX4 expression is linked to heightened resistance to ferroptosis in cancer cells. Moreover, the review discusses the implications of EMT-induced transcription factors such as Snail, Zeb1, and Twist in modulating the sensitivity of tumor cells to ferroptosis, thereby affecting drug resistance and cancer treatment outcomes. Targeting the ferroptosis pathway offers a promising therapeutic strategy, particularly for tumors resistant to conventional treatments. The induction of ferroptosis in these cells could potentially overcome drug resistance. However, translating these findings into clinical practice presents challenges, including understanding the precise mechanisms of ferroptosis induction, identifying predictive biomarkers, and optimizing combination therapies. The review underscores the need for further research to unravel the complex interactions between ferroptosis, EMT, and drug resistance in cancer. This could lead to the development of more effective, targeted cancer treatments, particularly for drug-resistant tumors, offering new hope in cancer therapeutics.
Collapse
Affiliation(s)
- Huiming Zhang
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Naifeng Chen
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Chenglong Ding
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Huinan Zhang
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| | - Dejiang Liu
- College of Biology and Agriculture, Jiamusi University, Jiamusi, China
| | - Shuang Liu
- School of Basic Medicine, Jiamusi University, Jiamusi, China
| |
Collapse
|
5
|
Song Y, Qu Y, Mao C, Zhang R, Jiang D, Sun X. Post-translational modifications of Keap1: the state of the art. Front Cell Dev Biol 2024; 11:1332049. [PMID: 38259518 PMCID: PMC10801156 DOI: 10.3389/fcell.2023.1332049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
The Keap1-Nrf2 signaling pathway plays a crucial role in cellular defense against oxidative stress-induced damage. Its activation entails the expression and transcriptional regulation of several proteins involved in detoxification and antioxidation processes within the organism. Keap1, serving as a pivotal transcriptional regulator within this pathway, exerts control over the activity of Nrf2. Various post-translational modifications (PTMs) of Keap1, such as alkylation, glycosylation, glutathiylation, S-sulfhydration, and other modifications, impact the binding affinity between Keap1 and Nrf2. Consequently, this leads to the accumulation of Nrf2 and its translocation to the nucleus, and subsequent activation of downstream antioxidant genes. Given the association between the Keap1-Nrf2 signaling pathway and various diseases such as cancer, neurodegenerative disorders, and diabetes, comprehending the post-translational modification of Keap1 not only deepens our understanding of Nrf2 signaling regulation but also contributes to the identification of novel drug targets and biomarkers. Consequently, this knowledge holds immense importance in the prevention and treatment of diseases induced by oxidative stress.
Collapse
Affiliation(s)
- Yunjia Song
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ying Qu
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Caiyun Mao
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Rong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Deyou Jiang
- Department of Typhoid, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xutao Sun
- Department of Synopsis of the Golden Chamber, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
6
|
Li L, Jia Q, Wang X, Wang Y, Wu C, Cong J, Ling J. Chaihu Shugan San promotes gastric motility in rats with functional dyspepsia by regulating Drp-1-mediated ICC mitophagy. PHARMACEUTICAL BIOLOGY 2023; 61:249-258. [PMID: 36655341 PMCID: PMC9858526 DOI: 10.1080/13880209.2023.2166966] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
CONTEXT Chaihu Shugan San (CHSGS) was effective in the treatment of functional dyspepsia (FD). OBJECTIVE To investigate the mechanism of CHSGS in FD through dynamin-related protein 1 (Drp-1)-mediated interstitial cells of cajal (ICC) mitophagy. MATERIALS AND METHODS Forty Sprague-Dawley (SD) rats were randomly divided into control, model, mdivi-1, mdivi-1 + CHSGS and CHSGS groups. Tail-clamping stimulation was used to establish the FD model. Mdivi-1 + CHSGS and CHSGS groups were given CHSGS aqueous solution (4.8 g/kg) by gavage twice a day. Mdivi-1 (25 mg/kg) was injected intraperitoneally once every other week for 4 w. Mitochondrial damage was observed by corresponding kits and related protein expressions were assessed by Immunofluorescence and (or) Western Blot. RESULTS Compared with the mean value of the control group, superoxide dismutase (SOD) and citrate synthase (CS) in the model group were decreased by 11% and 35%; malondialdehyde (MDA) and reactive oxygen species (ROS) were increased by 1.2- and 2.8-times; ckit fluorescence and protein expressions were decreased by 85% and 51%, co-localization expression of LC3 and voltage dependent anion channel 1 (VDAC1), Drp-1 and translocase of the outer mitochondrial membrane 20 (Tom20) were increased by 10.1- and 5.4-times; protein expressions of Drp-1, Beclin-1, and LC3 were increased by 0.5-, 1.4-, and 2.5-times whereas p62 was decreased by 43%. After mdivi-1 and (or) CHSGS intervention, the above situation has been improved. DISCUSSION AND CONCLUSION CHSGS could improve mitochondrial damage and promote gastric motility in FD rats by regulating Drp-1-mediated ICC mitophagy.
Collapse
Affiliation(s)
- Li Li
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Qingling Jia
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Xiangxiang Wang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yujiao Wang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Chenheng Wu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jun Cong
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Jianghong Ling
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- CONTACT Jianghong Ling Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai200021, People’s Republic of China
| |
Collapse
|
7
|
Saraiva C, Lopes-Nunes J, Esteves M, Santos T, Vale A, Cristóvão AC, Ferreira R, Bernardino L. CtBP Neuroprotective Role in Toxin-Based Parkinson's Disease Models: From Expression Pattern to Dopaminergic Survival. Mol Neurobiol 2023; 60:4246-4260. [PMID: 37060501 PMCID: PMC10293336 DOI: 10.1007/s12035-023-03331-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 02/07/2023] [Indexed: 04/16/2023]
Abstract
C-terminal binding proteins (CtBP) are transcriptional co-repressors regulating gene expression. CtBP promote neuronal survival through repression of pro-apoptotic genes, and may represent relevant targets for neurodegenerative disorders, such as Parkinson's disease (PD). Nevertheless, evidence of the role of CtBP1 and CtBP2 in neurodegeneration are scarce. Herein, we showed that CtBP1 and CtBP2 are expressed in neurons, dopaminergic neurons, astrocytes, and microglia in the substantia nigra (SN) and striatum of adult mice. Old mice showed a lower expression of CtBP1 in the SN and higher expression of CtPB2 in the SN and striatum compared with adult mice. In vivo models for PD (paraquat, MPTP, 6-OHDA) showed increased expression of CtBP1 in the SN and striatum while CtBP2 expression was increased in the striatum of paraquat-treated rats only. Moreover, an increased expression of both CtBP was found in a dopaminergic cell line (N27) exposed to 6-OHDA. In the 6-OHDA PD model, we found a dual effect using an unspecific ligand of CtBP, the 4-methylthio 2-oxobutyric acid (MTOB): higher concentrations (e.g. 2500 µM, 1000 µM) inhibited dopaminergic survival, while at 250 μM it counteracted cell death. In vitro, this latter protective role was absent after the siRNA silencing of CtBP1 or CtBP2. Altogether, this is the first report exploring the cellular and regional expression pattern of CtBP in the nigrostriatal pathway and the neuroprotective role in PD toxin-based models. CtBP could counteract dopaminergic cell death in the 6-OHDA PD model and, therefore, CtBP function and therapeutic potential in PD should be further explored.
Collapse
Affiliation(s)
- Cláudia Saraiva
- Brain Repair Group, Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Present Address: Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 Avenue Des Hauts-Fourneaux, Esch-Sur-Alzette, Luxembourg
| | - Jéssica Lopes-Nunes
- Brain Repair Group, Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Marta Esteves
- Brain Repair Group, Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Tiago Santos
- Brain Repair Group, Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Vale
- Brain Repair Group, Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Ana Clara Cristóvão
- Brain Repair Group, Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| | - Raquel Ferreira
- Brain Repair Group, Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
- Present Address: CEDOC, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria, 130, Lisboa, Portugal
| | - Liliana Bernardino
- Brain Repair Group, Health Sciences Research Center (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal
| |
Collapse
|
8
|
Özatik FY, Özatik O, Tekşen Y, Koçak H, Arı NS, Çengelli Ünel Ç. Dose-Dependent Effect of Hydrogen Sulfide in Cyclophosphamide-Induced Hepatotoxicity in Rats. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2023; 34:626-634. [PMID: 37162504 PMCID: PMC10441066 DOI: 10.5152/tjg.2023.22040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 09/12/2022] [Indexed: 05/11/2023]
Abstract
BACKGROUND Cyclophosphamide is a commonly used anticancer and immunosuppressive agent; however, hepatotoxicity is one of its severe toxicities. Hydrogen sulfide is a gaseous signaling molecule that plays crucial regulatory roles in various physiological functions. This study aimed to evaluate the hepatoprotective effect of hydrogen sulfide against cyclo phosp hamid e-ind uced hepatic damage in rats. METHODS Hepatotoxicity was induced by the single intraperitoneal administration of cyclophosphamide (200 mg/kg). Sprague-Dawley rats were treated by hydrogen sulfide donor, sodium hydrosulfide (25, 50, and 100 μmol/kg, intraperitoneal) 7 days before and 7 days after the administration of a single intraperitoneal injection of cyclophosphamide (200 mg/kg). Cyclo phosp hamide-ind uced hepatotoxicity was evaluated by serum and tissue biochemical and histopathological assessments. The levels of hydrogen sulfide, nitric oxide, cyclic guanosine monophosphate, interleukin 6, and interleukin 10 in liver homogenates were also determined by ELISA. One-way analysis of variance and Kruskal-Wallis tests were used as statistical analyses. RESULTS Cyclophosphamide increased liver function enzymes (alanine aminotransferase and aspartate aminotransferase), immunoreactivity to caspase-3 and Apaf-1, and proinflammatory cytokines. Cyclophosphamide also induced histopathological alterations including pycnotic nucleus with eosinophilic cytoplasm, increased sinusoidal dilatation, congestion, and edema. Hydrogen sulfide cotreatment significantly reduced cyclo phosp hamid e-ind uced inflammation, histological alterations, and apoptosis in the liver. 50 mg/kg sodium hydrosulfide was more effective against cyclo phosp hamid e-ind uced hepatotoxicity. CONCLUSION In conclusion, hydrogen sulfide with its anti-inflammatory and anti-apoptotic effects seems to be beneficial as an adjunct to cyclophosphamide treatment to reduce cyclo phosp hamid e-ind uced hepatotoxicity and thereby can be suggested as a promising agent to increase the therapeutic efficacy of cyclophosphamide.
Collapse
Affiliation(s)
- Fikriye Yasemin Özatik
- Department of Pharmacology, Kütahya Health Sciences University Faculty of Medicine, Kütahya, Turkey
| | - Orhan Özatik
- Department of Histology and Embryology, Kütahya Health Sciences University Faculty of Medicine, Kütahya, Turkey
| | - Yasemin Tekşen
- Department of Pharmacology, Kütahya Health Sciences University Faculty of Medicine, Kütahya, Turkey
| | - Havva Koçak
- Department of Medical Biochemistry, Kütahya Health Sciences University Faculty of Medicine, Kütahya, Turkey
| | - Neziha Senem Arı
- Department of Histology and Embryology, Kütahya Health Sciences University Faculty of Medicine, Kütahya, Turkey
| | - Çiğdem Çengelli Ünel
- Department of Pharmacology, Eskişehir Osmangazi University Faculty of Medicine, Eskişehir, Turkey
| |
Collapse
|
9
|
Liu F, Yuan L, Li L, Yang J, Liu J, Chen Y, Zhang J, Lu Y, Yuan Y, Cheng J. S-sulfhydration of SIRT3 combats BMSC senescence and ameliorates osteoporosis via stabilizing heterochromatic and mitochondrial homeostasis. Pharmacol Res 2023; 192:106788. [PMID: 37146925 DOI: 10.1016/j.phrs.2023.106788] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 04/23/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Senescence of bone marrow mesenchymal stem cells (BMSCs) is one of the leading causes of osteoporosis. SIRT3, an essential NAD-dependent histone deacetylase, is highly correlated with BMSC senescence-mediated bone degradation and mitochondrial/heterochromatic disturbance. S-sulfhydration of cysteine residues favorably enhances SIRT3 activity by forming persulfides. Nevertheless, the underlying molecular mechanism of SIRT3 S-sulfhydration on mitochondrial/heterochromatic homeostasis involved in BMSC senescence remains unknown. Here, we demonstrated that CBS and CSE, endogenous hydrogen sulfide synthases, are downregulated with BMSC senescence. Exogenous H2S donor NaHS-mediated SIRT3 augmentation rescued the senescent phenotypes of BMSCs. Conversely, SIRT3 deletion accelerated oxidative stress-induced BMSC senescence through mitochondrial dysfunction and the detachment of the heterochromatic protein H3K9me3 from the nuclear envelope protein Lamin B1. H2S-mediated SIRT3 S-sulfhydration modification rescued the disorganized heterochromatin and fragmented mitochondria induced by the S-sulfhydration inhibitor dithiothreitol, thus leading to elevated osteogenic capacity and preventing BMSC senescence. The antisenescence effect of S-sulfhydration modification on BMSCs was abolished when the CXXC sites of the SIRT3 zinc finger motif were mutated. In vivo, aged mice-derived BMSCs pretreated with NaHS were orthotopically transplanted to the ovariectomy-induced osteoporotic mice, and we proved that SIRT3 ameliorates bone loss by inhibiting BMSC senescence. Overall, our study for the first time indicates a novel role of SIRT3 S-sulfhydration in stabilizing heterochromatin and mitochondrial homeostasis in counteracting BMSC senescence, providing a potential target for the treatment of degenerative bone diseases.
Collapse
Affiliation(s)
- Fei Liu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Longhui Yuan
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lan Li
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingchao Yang
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingping Liu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Younan Chen
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhang
- Core Facility of West China Hospital, Sichuan University, Chengdu P.R. China
| | - Yanrong Lu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yujia Yuan
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Jingqiu Cheng
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Deng G, Muqadas M, Adlat S, Zheng H, Li G, Zhu P, Nasser MI. Protective Effect of Hydrogen Sulfide on Cerebral Ischemia-Reperfusion Injury. Cell Mol Neurobiol 2023; 43:15-25. [PMID: 35066714 PMCID: PMC11415178 DOI: 10.1007/s10571-021-01166-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/01/2021] [Indexed: 01/07/2023]
Abstract
The brain is the most sensitive organ to hypoxia in the human body. Hypoxia in the brain will lead to damage to local brain tissue. When the blood supply of ischemic brain tissue is restored, the damage will worsen, that is, cerebral ischemia-reperfusion injury. Hydrogen sulfide (H2S) is a gaseous signal molecule and a novel endogenous neuroregulator. Indeed, different concentrations of H2S have different effects on neurons. Low concentration of H2S can play an important protective role in cerebral ischemia-reperfusion injury by inducing anti-oxidative stress injury, inhibition of inflammatory response, inhibition of cell apoptosis, reduction of cerebrovascular endothelial cell injury, regulation of autophagy, and other ways, which provides a new idea for clinical diagnosis and treatment of related diseases. This review aims to report the recent research progress on the dual effect of H2S on brain tissue during cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Gang Deng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, People's Republic of China
- Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, 510080, People's Republic of China
| | - Masood Muqadas
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, People's Republic of China
- Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, 510080, People's Republic of China
| | - Salah Adlat
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, People's Republic of China
- Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, 510080, People's Republic of China
| | - Haiyun Zheng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, People's Republic of China
- Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, 510080, People's Republic of China
| | - Ge Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, People's Republic of China.
- Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, 510080, People's Republic of China.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, People's Republic of China.
- Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, 510080, People's Republic of China.
| | - M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, People's Republic of China.
- Guangdong Academy of Medical Sciences, 106 ZhongshanEr Road, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
11
|
Seckin Dinler B, Cetinkaya H, Secgin Z. The regulation of glutathione s-transferases by gibberellic acid application in salt treated maize leaves. PHYSIOLOGY AND MOLECULAR BIOLOGY OF PLANTS : AN INTERNATIONAL JOURNAL OF FUNCTIONAL PLANT BIOLOGY 2023; 29:69-85. [PMID: 36733837 PMCID: PMC9886772 DOI: 10.1007/s12298-022-01269-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 06/18/2023]
Abstract
Plant hormones and antioxidant system changes occur during plants' exposure to stress conditions. Although the interactions of some plant hormones (abscisic acid, salicylic acid, jasmonic acid, nitric oxide, and ethylene) with the glutathione s-transferase (GST) enzyme, which is one of the antioxidant enzymes, have already been reported, the influence of gibberellic acid (GA3) on this enzyme under saline conditions has not yet been reported. Plant material for the experiments was obtained from M14G144 cultivar of maize (Zea mays L.) plants grown as a soil culture in growth chambers at 22 °C, 65-70% moisture, 16-h light/8-h dark conditions, and with full strength Hoagland solution for 8 days under controlled growth conditions. Then, the plants were exposed to salt stress (350 mM NaCl and 100, 300, and 500 ppm GA3) simultaneously. In maize leaves, GA3 treatment alleviated the physiological parameters under salt stress. Specifically, the treatments with 100 and 500 ppm of GA3 were able to trigger GST enzyme and isoenzyme activities as well as hydrogen sulfide accumulation and anthocyanin content, although the lowest malondialdehyde, hydrogen peroxide, and superoxide radical content were under the treatment of 300 ppm of GA3. Besides this, GST gene expression levels were found to be upregulated between 1.5 and fourfold higher in all the plants treated with GA3 at different concentrations in proportion to salt stress. These results first indicated that the reason for the changes in GA3-treated plants was the stimulating role of this hormone to maintain GST regulation in maize plants. Supplementary Information The online version contains supplementary material available at 10.1007/s12298-022-01269-2.
Collapse
Affiliation(s)
- Burcu Seckin Dinler
- Department of Biology, Faculty of Arts and Sciences, Sinop University, Sinop, Turkey
| | - Hatice Cetinkaya
- Department of Biology, Faculty of Arts and Sciences, Sinop University, Sinop, Turkey
| | - Zafer Secgin
- Department of Agricultural Biotechnology, Ondokuz Mayıs University, Samsun, Turkey
| |
Collapse
|
12
|
Li J, Zhao M, Li J, Wang M, Zhao C. Combining fecal microbiome and metabolomics to reveal the disturbance of gut microbiota in liver injury and the therapeutic mechanism of shaoyao gancao decoction. Front Pharmacol 2022; 13:911356. [PMID: 36059945 PMCID: PMC9428823 DOI: 10.3389/fphar.2022.911356] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Chemical liver injury is closely related to gut microbiota and its metabolites. In this study, we combined 16S rRNA gene sequencing, 1H NMR-based fecal metabolomics and GC-MS to evaluate the changes in gut microbiota, fecal metabolites and Short-chain fatty acids (SCFAs) in CCl4-induced liver injury in Sprague-Dawley rats, and the therapeutic effect of Shaoyao Gancao Decoction (SGD). The results showed that CCl4-induced liver injury overexpressed CYP2E1, enhanced oxidative stress, decreased antioxidant enzymes (SOD, GSH), increased peroxidative products MDA and inflammatory responses (IL-6, TNF-α), which were ameliorated by SGD treatment. H&E staining showed that SGD could alleviate liver tissue lesions, which was confirmed by the recovered liver index, ALT and AST. Correlation network analysis indicated that liver injury led to a decrease in microbiota correlation, while SGD helped restore it. In addition, fecal metabolomic confirmed the PICRUSt results that liver injury caused disturbances in amino acid metabolism, which were modulated by SGD. Spearman’s analysis showed that liver injury disrupted ammonia transport, urea cycle, intestinal barrier and energy metabolism. Moreover, the levels of SCFAs were also decreased, and the abundance of Lachnoclostridium, Blautia, Lachnospiraceae_NK4A136_group, UCG-005 and Turicibacter associated with SCFAs were altered. However, all this can be alleviated by SGD. More importantly, pseudo germ-free rats demonstrated that the absence of gut microbiota aggravated liver injury and affected the efficacy of SGD. Taken together, we speculate that the gut microbiota has a protective role in the pathogenesis of liver injury, and has a positive significance for the efficacy of SGD. Moreover, SGD can treat liver injury by modulating gut microbiota and its metabolites and SCFAs. This provides useful evidence for the study of the pathogenesis of liver injury and the clinical application of SGD.
Collapse
Affiliation(s)
- Jingwei Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Min Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Jianming Li
- Guangxi University of Chinese Medicine, Nanning, China
| | - Miao Wang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
- *Correspondence: Miao Wang, ; Chunjie Zhao,
| | - Chunjie Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
- *Correspondence: Miao Wang, ; Chunjie Zhao,
| |
Collapse
|
13
|
Jia Q, Li L, Wang X, Wang Y, Jiang K, Yang K, Cong J, Cai G, Ling J. Hesperidin promotes gastric motility in rats with functional dyspepsia by regulating Drp1-mediated ICC mitophagy. Front Pharmacol 2022; 13:945624. [PMID: 36034863 PMCID: PMC9412972 DOI: 10.3389/fphar.2022.945624] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/05/2022] [Indexed: 11/22/2022] Open
Abstract
Hesperidin is one of the main active ingredients of Citrus aurantiumL. (Rutaceae) and tangerine peel, which have anti-inflammatory and antioxidant effects. In previous study, we found that gastric motility disorder in functional dyspepsia (FD) rats accompanied by excessive autophagy/mitochondrial swelling and even vacuolization in the interstitial cells of cajal (ICC), but the exact mechanism has not yet been investigated. Therefore, we used different doses of hesperidin (50 mg/kg, 100 mg/kg, and 200 mg/kg) to intervene in FD rats, and found that medium doses of hesperidin (100 mg/kg) significantly increased gastric motility in FD rats. Subsequently, FD rats were randomly divided into control group, model group, mdivi-1 group, mdivi-1+hesperidin group and hesperidin group, and mitochondrial division inhibitor (mdivi-1) was injected intraperitoneally to further investigate whether hesperidin could regulate dynamin-related protein 1 (Drp1)-mediated mitophagy in ICC to improve mitochondrial damage. The results showed that compared with the model group, the serum malondialdehyde (MDA) level decreased and the superoxide dismutase (SOD) level increased in the mdivi-1 and hesperidin groups (p < 0.001). Transmission electron microscopy (TEM) observed that the mitochondrial nuclear membrane was intact in gastric tissues with a clear internal cristae pattern, and autophagy lysosomes were rare. The co-localization expression of microtubule associated protein 1 light chain 3 (LC3) and voltage dependent anion channel 1 (VDAC1), Drp1 and translocase of the outer mitochondrial membrane 20 (Tom20) was significantly decreased (p < 0.001), the protein expression of mitochondrial Drp1, Beclin1 and LC3 were significantly decreased (p < 0.001), the protein expression of mitochondrial P62 and ckit in gastric tissue were significantly increased (p < 0.05, p < 0.001). The above situation was improved more significantly by the synergistic intervention of mdivi-1 and hesperidin. Therefore, hesperidin can improve mitochondrial damage and promote gastric motility in FD rats by regulating Drp1-mediated ICC mitophagy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Gan Cai
- *Correspondence: Gan Cai, ; Jianghong Ling,
| | | |
Collapse
|
14
|
Hydrogen Sulfide Ameliorated High Choline-Induced Cardiac Dysfunction by Inhibiting cGAS-STING-NLRP3 Inflammasome Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1392896. [PMID: 35910846 PMCID: PMC9337966 DOI: 10.1155/2022/1392896] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022]
Abstract
Although it is an essential nutrient, high choline intake directly or indirectly via its metabolite is associated with increased risk of cardiovascular disease, the mechanism of which remains to be elucidated. The present study was performed to investigate whether hydrogen sulfide (H2S) was involved in high choline-induced cardiac dysfunction and explore the potential mechanisms. We found that ejection fraction (EF) and fractional shortening (FS), the indicators of cardiac function measured by echocardiography, were significantly decreased in mice fed a diet containing 1.3% choline for 4 months as compared to the control, while applying 3,3-dimethyl-1-butanol (DMB) to suppress trimethylamine N-oxide (TMAO, a metabolite of choline) generation ameliorated the cardiac function. Subsequently, we found that feeding choline or TMAO significantly increased the protein levels of cyclic GMP-AMP (cGAMP) synthase (cGAS), stimulator of interferon genes (STING), NOD-like receptor protein 3 (NLRP3), caspase-1, and interleukin-1β (IL-1β) as compared to the control, which indicated the activation of cGAS-STING-NLRP3 inflammasome axis. Moreover, the protein expression of cystathionine γ-lyase (CSE), the main enzyme for H2S production in the cardiovascular system, was significantly increased after dietary supplementation with choline, but the plasma H2S levels were significantly decreased. To observe the effect of endogenous H2S, CSE knockout (KO) mice were used, and we found that the EF, FS, and plasma H2S levels in WT mice were significantly decreased after dietary supplementation with choline, while there was no difference between CSE KO + control and CSE KO + choline group. To observe the effect of exogenous H2S, mice were intraperitoneally injected with sodium hydrosulfide (NaHS, a H2S donor) for 4 months, and we found that NaHS improved the cardiac function and reduced the protein levels of cGAS, STING, NLRP3, caspase-1, and IL-1β in mice receiving dietary choline. In conclusion, our studies revealed that high choline diet decreased plasma H2S levels and induced cardiac dysfunction via cGAS-STING-NLRP3 inflammasome axis while H2S treatment could restore the cardiac function by inhibiting cGAS-STING-NLRP3 inflammasome axis.
Collapse
|
15
|
Deficiency in Inactive Rhomboid Protein2 (iRhom2) Alleviates Alcoholic Liver Fibrosis by Suppressing Inflammation and Oxidative Stress. Int J Mol Sci 2022; 23:ijms23147701. [PMID: 35887045 PMCID: PMC9317380 DOI: 10.3390/ijms23147701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/04/2022] [Accepted: 07/09/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic alcohol exposure can lead to liver pathology relating to inflammation and oxidative stress, which are two of the major factors in the incidence of liver fibrosis and even liver cancer. The underlying molecular mechanisms regarding hepatic lesions associated with alcohol are not fully understood. Considering that the recently identified iRhom2 is a key pathogenic mediator of inflammation, we performed in vitro and in vivo experiments to explore its regulatory role in alcohol-induced liver fibrosis. We found that iRhom2 knockout significantly inhibited alcohol-induced inflammatory responses in vitro, including elevated expressions of inflammatory cytokines (IL-1β, IL-6, IL-18, and TNF-α) and genes associated with inflammatory signaling pathways, such as TACE (tumor necrosis factor-alpha converting enzyme), TNFR1 (tumor necrosis factor receptor 1), and TNFR2, as well as the activation of NF-κB. The in vivo results confirmed that long-term alcohol exposure leads to hepatocyte damage and fibrous accumulation. In this pathological process, the expression of iRhom2 is promoted to activate the TACE/NF-κB signaling pathway, leading to inflammatory responses. Furthermore, the deletion of iRhom2 blocks the TACE/NF-κB signaling pathway and reduces liver damage and fibrosis caused by alcohol. Additionally, the activation of the JNK/Nrf2/HO-1 signaling pathway caused by alcohol exposure was also noted in vitro and in vivo. In the same way, knockout or deleting iRhom2 blocked the JNK/Nrf2/HO-1 signaling pathway to regulate the oxidative stress. Therefore, we contend that iRhom2 is a key regulator that promotes inflammatory responses and regulates oxidative stress in alcoholic liver fibrosis lesions. We posit that iRhom2 is potentially a new therapeutic target for alcoholic liver fibrosis.
Collapse
|
16
|
Alizadeh S, Anani-Sarab G, Amiri H, Hashemi M. Paraquat induced oxidative stress, DNA damage, and cytotoxicity in lymphocytes. Heliyon 2022; 8:e09895. [PMID: 35855999 PMCID: PMC9287805 DOI: 10.1016/j.heliyon.2022.e09895] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/26/2022] [Accepted: 07/01/2022] [Indexed: 11/26/2022] Open
Abstract
Paraquat (PQ) is a herbicide belonging to the group of bipyridylium salts. The objective of this study was to evaluate oxidative stress, DNA damage, and cytotoxicity induced by paraquat in peripheral lymphocyte cells in vivo as well as pathological changes in various tissues. For this purpose, 28 male Wistar rats in 6 different groups were poisoned by paraquat gavage and blood samples were taken from the hearts of rats after during the poisoning period. Oxidative stress, DNA damage, cell membrane integrity, serum lactate dehydrogenase, and cytotoxicity, were investigated by Ferric Reducing Antioxidant Potential (FRAP) test, alkaline comet assay, measuring serum lactate dehydrogenase (LDH), Hoechst staining and flow cytometry with propidium iodide (PI) respectively. The lung, kidney, and liver tissues were also examined pathologically. Paraquat caused dose-dependent DNA damage in peripheral lymphocyte cells and significant oxidative cell membrane damage. The most damage was caused by a single dose of 200 mg/kg b.w of paraquat by gavage. The gradual exposure to a dose of 300 mg/kg b.w of paraquat showed less damage, which could be due to the activation of the antioxidant defense mechanism. Paraquat induced oxidative stress. Paraquat increases serum lactate dehydrogenase. Oxidative stress Inducted by exposure to paraquat Inducted DNA damage.
Collapse
Affiliation(s)
- Soheila Alizadeh
- Environmental Health Engineering Research Center, Kerman University of Medical Sciences, Kerman, Iran.,Department of Environmental Health Engineering, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Anani-Sarab
- Medical Toxicology & Drug Abuse Research Center Birjand University of Medical Sciences, Birjand, Iran.,School of Allied Medical Sciences Birjand University of Medical Sciences, Birjand, Iran
| | - Hoda Amiri
- Environmental Health Engineering Research Center, Kerman University of Medical Sciences, Kerman, Iran.,Department of Environmental Health Engineering, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Hashemi
- Environmental Health Engineering Research Center, Kerman University of Medical Sciences, Kerman, Iran.,Department of Environmental Health Engineering, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran.,Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
17
|
Khalaf HM, Hafez SMNA, Abdalla AM, Welson NN, Abdelzaher WY, Abdelbaky FAF. Role of Platelet-activating factor and HO-1 in mediating the protective effect of rupatadine against 5-fluorouracil-induced hepatotoxicity in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:40190-40203. [PMID: 35119631 PMCID: PMC9120097 DOI: 10.1007/s11356-022-18899-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 01/23/2022] [Indexed: 05/06/2023]
Abstract
5-fluorouracil (5-FU) is a widely used chemotherapeutic drug, but its hepatotoxicity challenges its clinical use. Thus, searching for a hepatoprotective agent is highly required to prevent the accompanied hepatic hazards. The current study aimed to investigate the potential benefit and mechanisms of action of rupatadine (RU), a Platelet-activating factor (PAF) antagonist, in the prevention of 5-FU-related hepatotoxicity in rats. Hepatotoxicity was developed in male albino rats by a single 5-FU (150 mg/kg) intra-peritoneal injection on the 7th day of the experiment. RU (3 mg/kg/day) was orally administrated to the rodents for 10 days. Hepatic toxicity was assessed by measuring both liver and body weights, serum alanine aminotransferase and aspartate aminotransferase (ALT and AST), hepatic oxidative stress parameters (malondialdehyde (MDA), nitric oxide levels (NOx), reduced glutathione (GSH), superoxide dismutase (SOD)), and heme oxygenase-1 (HO-1). Inflammatory markers expressions (inducible nitric oxide synthase (iNOS), tumor necrosis factor-alpha (TNFα), interleukins; IL-1B, IL-6), the apoptotic marker (caspase-3), and PAF were measured in the hepatic tissue. 5-FU-induced hepatotoxicity was proved by the biochemical along with histopathological assessments. RU ameliorated 5-FU-induced liver damage as proved by the improved serum ALT, AST, and hepatic oxidative stress parameters, the attenuated expression of hepatic pro-inflammatory cytokines and PAF, and the up-regulation of HO-1. Therefore, it can be concluded that RU pretreatment exerted a hepatoprotective effect against 5-FU-induced liver damage through both its powerful anti-inflammatory, antioxidant, and anti-apoptotic effect.
Collapse
Affiliation(s)
| | | | | | - Nermeen N. Welson
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | | | | |
Collapse
|
18
|
Abdel-Latif R, Heeba GH, Hassanin SO, Waz S, Amin A. TLRs-JNK/ NF-κB Pathway Underlies the Protective Effect of the Sulfide Salt Against Liver Toxicity. Front Pharmacol 2022; 13:850066. [PMID: 35517830 PMCID: PMC9065287 DOI: 10.3389/fphar.2022.850066] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022] Open
Abstract
Hydrogen sulfide (H2S) is an endogenously gas transmitter signaling molecule with known antioxidant, anti-inflammatory, and cytoprotective properties. Although accumulating evidence shows the therapeutic potential of H2S in various hepatic diseases, its role in cyclophosphamide (CP)-induced hepatotoxicity remains elusive. The present study was undertaken to investigate the impact of endogenous and exogenous H2S on toll-like receptors (TLRs)-mediated inflammatory response and apoptosis in CP-induced hepatotoxicity. Either an H2S donor (NaHS (100 μM/kg) or an H2S blocker [dl-propargylglycine (PAG) (30 mg/kg, i. p.)], was administered for 10 days before a single ip injection of CP (200 mg/kg). NaHS attenuated conferred hepatoprotection against CP-induced toxicity, significantly decreasing serum hepatic function tests and improving hepatic histopathology. Additionally, NaHS-treated rats exhibited antioxidant activity in liver tissues compared with the CP group. The upregulated hepatic levels of TLR2/4 and their downstream signaling molecules including c-Jun N-terminal kinase (JNK) and nuclear factor-kappa B (NF-κB) were also suppressed by NaHS protective treatment. NaHS showed anti-inflammatory and antiapoptotic effects; reducing hepatic level tumor necrosis factor-alpha (TNF-α) and caspase-3 expression. Interestingly, the cytotoxic events induced in CP-treated rats were not significantly altered upon the blocking of endogenous H2S. Taken together, the present study suggested that exogenously applied H2S rather than the endogenously generated H2S, displayed a hepatoprotective effect against CP-induced hepatotoxicity that might be mediated by TLRs-JNK/NF-κB pathways.
Collapse
Affiliation(s)
- Rania Abdel-Latif
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Gehan Hussein Heeba
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Soha Osama Hassanin
- Department of Biochemistry, Faculty of Pharmacy, MTI University, Cairo, Egypt
| | - Shaimaa Waz
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Amr Amin
- The College, The University of Chicago, Chicago, IL, United States.,Department of Biology, UAE University, Al Ain, United Arab Emirates
| |
Collapse
|
19
|
Wu X, Liu S, Zhu H, Ma Z, Dai X, Liu W. Scavenging ROS to Alleviate Acute Liver Injury by ZnO-NiO@COOH. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2103982. [PMID: 35138033 PMCID: PMC9008431 DOI: 10.1002/advs.202103982] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/14/2022] [Indexed: 05/06/2023]
Abstract
Currently, the incidence of acute liver injury (ALI) is increasing year by year, and infection with coronavirus disease 2019 (COVID-19) can also induce ALI, but there are still no targeted therapeutic drugs. ZnO-NiO particles is mainly used to clean up reactive oxygen species (ROS) in industrial wastewater, and it is insoluble in water. Its excellent properties are discovered and improved by adding shuttle-based bonds to make it more water-soluble. ZnO-NiO@COOH particles are synthetically applied to treat ALI. The p-n junction in ZnO-NiO@COOH increases the surface area and active sites, thereby creating large numbers of oxygen vacancies, which can quickly adsorb ROS. The content in tissues and serum levels of L-glutathione (GSH) and the GSH/oxidized GSH ratio are measured to assess the capacity of ZnO-NiO@COOH particles to absorb ROS. The ZnO-NiO@COOH particles significantly reduce the expression levels of inflammatory factors (i.e., IL-1, IL-6, and TNF-α), macrophage infiltration, and granulocyte activation. ZnO-NiO@COOH rapidly adsorb ROS in a short period of time to block the generation of inflammatory storms and gain time for the follow-up treatment of ALI, which has important clinical significance.
Collapse
Affiliation(s)
- Xuan Wu
- Central Laboratory and Department of Laboratory MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200070China
| | - Shiyu Liu
- State Key Laboratory of Pollution Control and Resource ReuseSchool of Environmental Science and EngineeringTongji UniversityShanghai200092China
- Shanghai Institute of Pollution Control and Ecological SecurityShanghai200092China
| | - Huanhuan Zhu
- Central Laboratory and Department of Laboratory MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200070China
| | - Zili Ma
- Central Laboratory and Department of Laboratory MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200070China
| | - Xiaohu Dai
- State Key Laboratory of Pollution Control and Resource ReuseSchool of Environmental Science and EngineeringTongji UniversityShanghai200092China
- Shanghai Institute of Pollution Control and Ecological SecurityShanghai200092China
| | - Weiwei Liu
- Central Laboratory and Department of Laboratory MedicineShanghai Tenth People's HospitalSchool of MedicineTongji UniversityShanghai200070China
- Department of Laboratory MedicineLonghua HospitalShanghai University of Traditional Chinese MedicineShanghai200032China
| |
Collapse
|
20
|
A hydrogen sulphide-responsive and depleting nanoplatform for cancer photodynamic therapy. Nat Commun 2022; 13:1685. [PMID: 35354794 PMCID: PMC8967875 DOI: 10.1038/s41467-022-29284-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/07/2022] [Indexed: 12/23/2022] Open
Abstract
Hydrogen sulfide (H2S) as an important biological gasotransmitter plays a pivotal role in many physiological and pathological processes. The sensitive and quantitative detection of H2S level is therefore crucial for precise diagnosis and prognosis evaluation of various diseases but remains a huge challenge due to the lack of accurate and reliable analytical methods in vivo. In this work, we report a smart, H2S-responsive and depleting nanoplatform (ZNNPs) for quantitative and real-time imaging of endogenous H2S for early diagnosis and treatment of H2S-associated diseases. We show that ZNNPs exhibit unexpected NIR conversion (F1070 → F720) and ratiometric photoacoustic (PA680/PA900) signal responsiveness towards H2S, allowing for sensitive and quantitative visualization of H2S in acute hepatotoxicity, cerebral hemorrhage model as well as colorectal tumors in living mice. ZNNPs@FA simultaneously scavenges the mitochondrial H2S in tumors leading to significant ATP reduction and severe mitochondrial damage, together with the activated photodynamic effect, resulting in efficient suppression of colorectal tumor growth in mice. We believe that this platform may provide a powerful tool for studying the vital impacts of H2S in related diseases.
Collapse
|
21
|
Qiu Y, Shen J, Jiang W, Yang Y, Liu X, Zeng Y. Sphingosine 1-phosphate and its regulatory role in vascular endothelial cells. Histol Histopathol 2022; 37:213-225. [PMID: 35118637 DOI: 10.14670/hh-18-428] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive metabolite of sphingomyelin. S1P activates a series of signaling cascades by acting on its receptors S1PR1-3 on endothelial cells (ECs), which plays an important role in endothelial barrier maintenance, anti-inflammation, antioxidant and angiogenesis, and thus is considered as a potential therapeutic biomarker for ischemic stroke, sepsis, idiopathic pulmonary fibrosis, cancers, type 2 diabetes and cardiovascular diseases. We presently review the levels of S1P in those vascular and vascular-related diseases. Plasma S1P levels were reduced in various inflammation-related diseases such as atherosclerosis and sepsis, but were increased in other diseases including type 2 diabetes, neurodegeneration, cerebrovascular damages such as acute ischemic stroke, Alzheimer's disease, vascular dementia, angina, heart failure, idiopathic pulmonary fibrosis, community-acquired pneumonia, and hepatocellular carcinoma. Then, we highlighted the molecular mechanism by which S1P regulated EC biology including vascular development and angiogenesis, inflammation, permeability, and production of reactive oxygen species (ROS), nitric oxide (NO) and hydrogen sulfide (H₂S), which might provide new ways for exploring the pathogenesis and implementing individualized therapy strategies for those diseases.
Collapse
Affiliation(s)
- Yan Qiu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Junyi Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yi Yang
- Department of Orthopeadics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
22
|
GPA Peptide Attenuates Sepsis-Induced Acute Lung Injury in Mice via Inhibiting Oxidative Stress and Pyroptosis of Alveolar Macrophage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:5589472. [PMID: 34992715 PMCID: PMC8727129 DOI: 10.1155/2021/5589472] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/03/2021] [Accepted: 12/02/2021] [Indexed: 02/08/2023]
Abstract
Acute lung injury (ALI) has been known to be a devastating form of respiratory infection and an important contributor to mortality in intensive care, due to its lacking of effective treatment. Inflammation, oxidative stress, and pyroptosis are associated with multiple kinds of inflammatory diseases such as ALI. It is commonly accepted that Gly-Pro-Ala (GPA) peptide regulates oxidative stress and pyroptosis in different kinds of inflammatory diseases. Our study is aimed at exploring the regulatory function and protective effects of GPA peptides on ALI. In the current study, the cecal ligation and puncture (CLP) technique was used to evoke sepsis in mice, and GPA peptide was administered intraperitoneally with different concentrations (50, 100, and 150 mg/kg) after CLP. Histopathological changes and the ratio of wet-to-dry in lung were recorded and analyzed. We also investigated the level of oxidative stress, inflammation, and pyroptosis. Results showed that GPA peptide significantly ameliorated CLP-stimulated lung tissue injury, impeded proinflammatory cytokine release, and reduced inflammatory cell infiltration. Additionally, GPA peptide suppressed oxidative stress and caspase-1-dependent pyroptosis in alveolar macrophages. Furthermore, our study showed that the GPA peptide prevents alveolar macrophage from undergoing pyroptosis by attenuating ROS. In conclusion, results demonstrated that GPA peptide has protective effects in CLP-stimulated ALI by inhibiting oxidative stress as well as pyroptosis of alveolar macrophage.
Collapse
|
23
|
Kalous KS, Wynia-Smith SL, Smith BC. Sirtuin Oxidative Post-translational Modifications. Front Physiol 2021; 12:763417. [PMID: 34899389 PMCID: PMC8652059 DOI: 10.3389/fphys.2021.763417] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
Increased sirtuin deacylase activity is correlated with increased lifespan and healthspan in eukaryotes. Conversely, decreased sirtuin deacylase activity is correlated with increased susceptibility to aging-related diseases. However, the mechanisms leading to decreased sirtuin activity during aging are poorly understood. Recent work has shown that oxidative post-translational modification by reactive oxygen (ROS) or nitrogen (RNS) species results in inhibition of sirtuin deacylase activity through cysteine nitrosation, glutathionylation, sulfenylation, and sulfhydration as well as tyrosine nitration. The prevalence of ROS/RNS (e.g., nitric oxide, S-nitrosoglutathione, hydrogen peroxide, oxidized glutathione, and peroxynitrite) is increased during inflammation and as a result of electron transport chain dysfunction. With age, cellular production of ROS/RNS increases; thus, cellular oxidants may serve as a causal link between loss of sirtuin activity and aging-related disease development. Therefore, the prevention of inhibitory oxidative modification may represent a novel means to increase sirtuin activity during aging. In this review, we explore the role of cellular oxidants in inhibiting individual sirtuin human isoform deacylase activity and clarify the relevance of ROS/RNS as regulatory molecules of sirtuin deacylase activity in the context of health and disease.
Collapse
Affiliation(s)
- Kelsey S Kalous
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Sarah L Wynia-Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
24
|
Anthrahydroquinone-2-6-disulfonate is a novel, powerful antidote for paraquat poisoning. Sci Rep 2021; 11:20159. [PMID: 34635711 PMCID: PMC8505516 DOI: 10.1038/s41598-021-99591-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/29/2021] [Indexed: 12/19/2022] Open
Abstract
Paraquat (PQ) is a widely used fast-acting pyridine herbicide. Accidental ingestion or self-administration via various routes can cause severe organ damage. Currently, no effective antidote is available commercially, and the mortality rate of poisoned patients is exceptionally high. Here, the efficacy of anthrahydroquinone-2-6-disulfonate (AH2QDS) was observed in treating PQ poisoning by constructing in vivo and ex vivo models. We then explored the detoxification mechanism of AH2QDS. We demonstrated that, in a rat model, the PQ concentration in the PQ + AH2QDS group significantly decreased compared to the PQ only group. Additionally, AH2QDS protected the mitochondria of rats and A549 cells and decreased oxidative stress damage, thus improving animal survival and cell viability. Finally, the differentially expressed genes were analysed in the PQ + AH2QDS group and the PQ group by NextGen sequencing, and we verified that Nrf2's expression in the PQ + AH2QDS group was significantly higher than that in the PQ group. Our work identified that AH2QDS can detoxify PQ by reducing PQ uptake and protecting mitochondria while enhancing the body's antioxidant activity.
Collapse
|
25
|
Hydrogen Sulfide Attenuates Angiotensin II-Induced Cardiac Fibroblast Proliferation and Transverse Aortic Constriction-Induced Myocardial Fibrosis through Oxidative Stress Inhibition via Sirtuin 3. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9925771. [PMID: 34603602 PMCID: PMC8486544 DOI: 10.1155/2021/9925771] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/30/2021] [Accepted: 08/07/2021] [Indexed: 12/12/2022]
Abstract
Sirtuin 3 (SIRT3) is critical in mitochondrial function and oxidative stress. Our present study investigates whether hydrogen sulfide (H2S) attenuated myocardial fibrosis and explores the possible role of SIRT3 on the protective effects. Neonatal rat cardiac fibroblasts were pretreated with NaHS followed by angiotensin II (Ang II) stimulation. SIRT3 was knocked down with siRNA technology. SIRT3 promoter activity and expression, as well as mitochondrial function, were measured. Male wild-type (WT) and SIRT3 knockout (KO) mice were intraperitoneally injected with NaHS followed by transverse aortic constriction (TAC). Myocardium sections were stained with Sirius red. Hydroxyproline content, collagen I and collagen III, α-smooth muscle actin (α-SMA), and dynamin-related protein 1 (DRP1) expression were measured both in vitro and in vivo. We found that NaHS enhanced SIRT3 promoter activity and increased SIRT3 mRNA expression. NaHS inhibited cell proliferation and hydroxyproline secretion, decreased collagen I, collagen III, α-SMA, and DRP1 expression, alleviated oxidative stress, and improved mitochondrial respiration function and membrane potential in Ang II-stimulated cardiac fibroblasts, which were unavailable after SIRT3 was silenced. In vivo, NaHS reduced hydroxyproline content, ameliorated perivascular and interstitial collagen deposition, and inhibited collagen I, collagen III, and DRP1 expression in the myocardium of WT mice but not SIRT3 KO mice with TAC. Altogether, NaHS attenuated myocardial fibrosis through oxidative stress inhibition via a SIRT3-dependent manner.
Collapse
|
26
|
Cano M, Datta S, Wang L, Liu T, Flores‐Bellver M, Sachdeva M, Sinha D, Handa JT. Nrf2 deficiency decreases NADPH from impaired IDH shuttle and pentose phosphate pathway in retinal pigmented epithelial cells to magnify oxidative stress-induced mitochondrial dysfunction. Aging Cell 2021; 20:e13444. [PMID: 34313391 PMCID: PMC8373343 DOI: 10.1111/acel.13444] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/26/2021] [Accepted: 07/08/2021] [Indexed: 12/14/2022] Open
Abstract
The nuclear factor‐erythroid 2‐related factor‐2 (Nrf2), a major antioxidant transcription factor, is decreased in several age‐related diseases including age‐related macular degeneration (AMD), the most common cause of blindness among the elderly in western society. Since Nrf2’s mito‐protective response is understudied, we investigated its antioxidant response on mitochondria. Control and Nrf2‐deficient retinal pigmented epithelial (RPE) cells were compared after treating with cigarette smoke extract (CSE). Mitochondrial antioxidant abundance and reactive oxygen species (ROS) were quantified. Mitochondrial function was assessed by TMRM assay, NADPH, electron transport chain activity, and Seahorse. Results were corroborated in Nrf2−/− mice and relevance to AMD was provided by immunohistochemistry of human globes. CSE induced mitochondrial ROS to impair mitochondrial function. H2O2 increase in particular, was magnified by Nrf2 deficiency, and corresponded with exaggerated mitochondrial dysfunction. While Nrf2 did not affect mitochondrial antioxidant abundance, oxidized PRX3 was magnified by Nrf2 deficiency due to decreased NADPH from decreased expression of IDH2 and pentose phosphate pathway (PPP) genes. With severe CSE stress, intrinsic apoptosis was activated to increase cell death. PPP component TALDO1 immunolabeling was decreased in dysmorphic RPE of human AMD globes. Despite limited regulation of mitochondrial antioxidant expression, Nrf2 influences PPP and IDH shuttle activity that indirectly supplies NADPH for the TRX2 system. These results provide insight into how Nrf2 deficiency impacts the mitochondrial antioxidant response, and its role in AMD pathobiology.
Collapse
Affiliation(s)
- Marisol Cano
- Wilmer Eye Institute Johns Hopkins School of Medicine Baltimore MD USA
| | - Sayantan Datta
- Wilmer Eye Institute Johns Hopkins School of Medicine Baltimore MD USA
| | - Lei Wang
- Wilmer Eye Institute Johns Hopkins School of Medicine Baltimore MD USA
| | - Tongyun Liu
- Wilmer Eye Institute Johns Hopkins School of Medicine Baltimore MD USA
| | | | - Mira Sachdeva
- Wilmer Eye Institute Johns Hopkins School of Medicine Baltimore MD USA
| | - Debasish Sinha
- Department of Ophthalmology University of Pittsburgh School of Medicine Baltimore MD USA
| | - James T. Handa
- Wilmer Eye Institute Johns Hopkins School of Medicine Baltimore MD USA
| |
Collapse
|
27
|
Huang Y, Wang G, Zhou Z, Tang Z, Zhang N, Zhu X, Ni X. Endogenous Hydrogen Sulfide Is an Important Factor in Maintaining Arterial Oxygen Saturation. Front Pharmacol 2021; 12:677110. [PMID: 34135757 PMCID: PMC8200772 DOI: 10.3389/fphar.2021.677110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 05/18/2021] [Indexed: 12/30/2022] Open
Abstract
The gasotransmitter H2S is involved in various physiological and pathophysiological processes. The aim of this study was to investigate the physiological functions of H2S in the lungs. In the model of mouse with genetic deficiency in a H2S natural synthesis enzyme cystathionine-γ-lyase (CSE), we found that arterial oxygen saturation (SaO2) was decreased compared with wild type mice. Hypoxyprobe test showed that mild hypoxia occurred in the tissues of heart, lungs and kidneys in Cse-/- mice. H2S donor GYY4137 treatment increased SaO2 and ameliorated hypoxia state in cardiac and renal tissues. Further, we revealed that lung blood perfusion and airway responsiveness were not linked to reduced SaO2 level. Lung injury was found in Cse-/- mice as evidenced by alveolar wall thickening, diffuse interstitial edema and leukocyte infiltration in pulmonary tissues. IL-8, IL-1β, and TNF-α levels were markedly increased and oxidative stress levels were also significantly higher with increased levels of the pro-oxidative biomarker, MDA, decreased levels of the anti-oxidative biomarkers, T-AOC and GSH/GSSG, and reduced superoxide dismutase (SOD) activity in lung tissues of Cse-/- mice compared with those of wild type mice. GYY4137 treatment ameliorated lung injury and suppressed inflammatory state and oxidative stress in lung tissues of Cse-/- mice. A decrease in SaO2 was found in normal mice under hypoxia. These mice displayed lung injury as evidenced by alveolar wall thickening, interstitial edema and leukocyte infiltration. Increased levels of inflammatory cytokines and oxidative stress were also found in lung tissues of the mice with hypoxia insult. GYY4137 treatment increased SaO2 and ameliorated lung injury, inflammation and oxidative stress. Our data indicate that endogenous H2S is an important factor in maintaining normal SaO2 by preventing oxidative stress and inflammation in the lungs.
Collapse
Affiliation(s)
- Yan Huang
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China.,Reproductive medicine center, Department of obstetrics and Gynecology, General Hospital of Southern Theater Command, Guangzhou, China
| | - Gang Wang
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhan Zhou
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China
| | - Zhengshan Tang
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China
| | - Ningning Zhang
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Xiaoyan Zhu
- Department of Physiology, Second Military Medical University, Shanghai, China
| | - Xin Ni
- National Clinical Research Center for Geriatric Disorders and Research Center for Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, China.,Department of Physiology, Second Military Medical University, Shanghai, China
| |
Collapse
|
28
|
NaHS Alleviated Cell Apoptosis and Mitochondrial Dysfunction in Remote Lung Tissue after Renal Ischemia and Reperfusion via Nrf2 Activation-Mediated NLRP3 Pathway Inhibition. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5598869. [PMID: 33954183 PMCID: PMC8064776 DOI: 10.1155/2021/5598869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/27/2021] [Accepted: 04/04/2021] [Indexed: 12/22/2022]
Abstract
Objective Acute kidney injury (AKI) is a common and severe complication in critically ill patients, often caused by renal ischemia-reperfusion (RIR). Previous studies have confirmed that lung injury, rather than renal injury, is one of the leading causes of AKI-induced death. The pathophysiological mechanisms of acute lung injury (ALI) resulting from AKI are very complex and remain unclear. In the present study, we aimed to explore the protective effects and potential mechanism of sodium hydrosulfide (NaHS) on lung injury in RIR mice. Methods The RIR model was established in wild-type and Nrf2−/− mice. Different groups of mice were treated with NaHS and MCC950. Lung tissues were harvested to detect lung injury, mitochondrial function, cell apoptosis, the NLRP3 inflammasome, and Nrf2 pathway-related molecules. Results RIR led to a deterioration in lung histology, the wet/dry weight ratio, PaO2/FiO2, and mitochondrial function, in addition to stimulating the activation of the NLRP3 and Nrf2 pathways. MCC950 alleviated mitochondrial dysfunction, lung apoptosis, and histology injury in the lungs after RIR. NaHS treatment markedly improved the lung histological scores, the wet/dry weight ratio, bronchoalveolar lavage fluid (BALF) cell counts, BALF neutrophil counts, BALF neutrophil elastase activity, BALF protein concentration, PaO2/FiO2, mitochondrial morphology, the red/green fluorescence intensity that indicates changes in mitochondrial membrane potential, respiratory control rate (RCR), ATP, reactive oxygen species (ROS) release, and cell apoptosis via Nrf2-mediated NLRP3 pathway inhibition. Conclusion NaHS protected against RIR-induced lung injury, mitochondrial dysfunction, and inflammation, which is associated with Nrf2 activation-mediated NLRP3 pathway inhibition.
Collapse
|
29
|
Li H, Yin Y, Liu J, Lu B, Wan H, Yang L, Wang W, Li R. Hydrogen-rich water attenuates the radiotoxicity induced by tritium exposure in vitro and in vivo. JOURNAL OF RADIATION RESEARCH 2021; 62:34-45. [PMID: 33231266 PMCID: PMC7779358 DOI: 10.1093/jrr/rraa104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 09/22/2020] [Indexed: 05/10/2023]
Abstract
Radionuclide tritium is widely used in the nuclear energy production industry and creates a threat to human health through radiation exposure. Herein, the radioactive elimination and radioprotective effect of hydrogen-rich water (HRW), a potential antioxidant with various medical applications, on tritiated water (HTO) exposure, was studied in vitro and in vivo. Results showed that intragastric administration of HRW effectively promoted the elimination of urinary tritium, decreased the level of serum tritium and tissue-bound tritium (OBT), and attenuated the genetic damage of blood cells in mice exposed to HTO (18.5 MBq/kg). Pretreatment with HRW effectively reduces tritium accumulation in HTO-treated human blood B lymphocyte AHH-1 cells. In addition, the anti-oxidative properties of HRW could attenuate the increased intracellular ROS (such as O2•-, •OH and ONOO-), resulting in reversing the exhaustion of cellular endogenous antioxidants (reduced GSH and SOD), decreasing lipid peroxidation (MDA), relieving DNA oxidative damage, and depressing cell apoptosis and cytotoxicity induced by HTO exposure. In conclusion, HRW is expected to be an effective radioactive elimination agent through the competition effect of isotope exchange or a radioprotective agent by scavenging free radicals induced by HTO exposure.
Collapse
Affiliation(s)
- Hong Li
- Centre for Diseases Prevention and Control of Eastern Theater, Nanjing 210002, China
- State Key Laboratory of Trauma Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Yaru Yin
- State Key Laboratory of Trauma Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Jing Liu
- State Key Laboratory of Trauma Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Binghui Lu
- State Key Laboratory of Trauma Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Huimin Wan
- State Key Laboratory of Trauma Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Luxun Yang
- State Key Laboratory of Trauma Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing 400038, China
| | - Weidong Wang
- Department of Radiation Oncology, Sichuan Cancer Hospital, Chengdu 610041, China
| | - Rong Li
- State Key Laboratory of Trauma Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University, Chongqing 400038, China
| |
Collapse
|
30
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Zarrin V, Moghadam ER, Hashemi F, Makvandi P, Samarghandian S, Khan H, Hashemi F, Najafi M, Mirzaei H. Toward Regulatory Effects of Curcumin on Transforming Growth Factor-Beta Across Different Diseases: A Review. Front Pharmacol 2020; 11:585413. [PMID: 33381035 PMCID: PMC7767860 DOI: 10.3389/fphar.2020.585413] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Immune response, proliferation, migration and angiogenesis are juts a few of cellular events that are regulated by transforming growth factor-β (TGF-β) in cells. A number of studies have documented that TGF-β undergoes abnormal expression in different diseases, e.g., diabetes, cancer, fibrosis, asthma, arthritis, among others. This has led to great fascination into this signaling pathway and developing agents with modulatory impact on TGF-β. Curcumin, a natural-based compound, is obtained from rhizome and roots of turmeric plant. It has a number of pharmacological activities including antioxidant, anti-inflammatory, anti-tumor, anti-diabetes and so on. Noteworthy, it has been demonstrated that curcumin affects different molecular signaling pathways such as Wnt/β-catenin, Nrf2, AMPK, mitogen-activated protein kinase and so on. In the present review, we evaluate the potential of curcumin in regulation of TGF-β signaling pathway to corelate it with therapeutic impacts of curcumin. By modulation of TGF-β (both upregulation and down-regulation), curcumin ameliorates fibrosis, neurological disorders, liver disease, diabetes and asthma. Besides, curcumin targets TGF-β signaling pathway which is capable of suppressing proliferation of tumor cells and invading cancer cells.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Istanbul, Turkey
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Vahideh Zarrin
- Laboratory for Stem Cell Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Pooyan Makvandi
- Centre for Micro-BioRobotics, Istituto Italiano di Tecnologia, Pisa, Italy
| | | | - Haroon Khan
- Student Research Committee, Department of Physiotherapy, Faculty of Rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fardin Hashemi
- Medical Technology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
31
|
Li L, Liu Y, Wang Q, Wang Z, Cui L, Xu Y, Guan K. Levels of nasal exhaled hydrogen sulfide in the general population and allergic rhinitis patients. J Clin Lab Anal 2020; 35:e23678. [PMID: 33615571 PMCID: PMC7957977 DOI: 10.1002/jcla.23678] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/08/2020] [Accepted: 11/22/2020] [Indexed: 12/11/2022] Open
Abstract
Background Objective measures used for the differential diagnosis and severity assessment of allergic rhinitis (AR) are still lacking. The involvement of hydrogen sulfide (H2S) in the development of AR indicates that nasal exhaled H2S (NeH2S) has potential as a biomarker to be used in AR patients. This study aimed to evaluate the application value of NeH2S measurement in the diagnosis and assessment of AR. Methods This study was a multi‐center cross‐sectional survey conducted in Northwestern China. Demographic information collection and rhinitis assessment were completed through questionnaires. The level of NeH2S and serum immunoglobulin E were measured. Results The level of NeH2S in general population ranged from 0 to 35 ppb, with a median value of 2 ppb. The NeH2S levels in seasonal allergic rhinitis (SAR) patients were significantly lower than those in general population (2 [1, 2.75] vs. 2 [2, 3] ppb; p = .023), and the NeH2S value of the SAR group tended to be lower than that of the non‐allergic rhinitis (NAR) group (2 [1, 2.75] vs. 2 [2, 3] ppb; p = .094). The subgroup of AR patients with symptoms lasting longer than 2 weeks per month had a lower NeH2S level compared with the subgroup of patients with symptoms lasting less than 2 weeks per month (2 [1, 2] vs. 2 [2, 3] ppb; p = .015). Conclusion This study described the distribution range of NeH2S levels in the general population. Further study with larger sample size was needed to clarify the relationship between NeH2S level and AR.
Collapse
Affiliation(s)
- Lisha Li
- Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Yonglin Liu
- Department of Pediatrics, Shenmu Hospital, Shenmu, China
| | - Qiang Wang
- Department of Orthopedics, Shenmu Hospital, Shenmu, China
| | - Zixi Wang
- Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Le Cui
- Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Yingyang Xu
- Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Kai Guan
- Beijing Key Laboratory of Precision Medicine for Diagnosis and Treatment on Allergic Diseases, Department of Allergy, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| |
Collapse
|
32
|
Wu D, Zhong P, Wang Y, Zhang Q, Li J, Liu Z, Ji A, Li Y. Hydrogen Sulfide Attenuates High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease by Inhibiting Apoptosis and Promoting Autophagy via Reactive Oxygen Species/Phosphatidylinositol 3-Kinase/AKT/Mammalian Target of Rapamycin Signaling Pathway. Front Pharmacol 2020; 11:585860. [PMID: 33390956 PMCID: PMC7774297 DOI: 10.3389/fphar.2020.585860] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common chronic liver disease worldwide. Hydrogen sulfide (H2S) is involved in a wide range of physiological and pathological processes. Nevertheless, the mechanism of action of H2S in NAFLD development has not been fully clarified. Here, the reduced level of H2S was observed in liver cells treated with oleic acid (OA). Administration of H2S increased the proliferation of OA-treated cells. The results showed that H2S decreased apoptosis and promoted autophagy through reactive oxygen species (ROS)-mediated phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) cascade in OA-treated cells. In addition, administration of H2S relieved high-fat diet (HFD)-induced NAFLD via inhibition of apoptosis and promotion of autophagy. These findings suggest that H2S could ameliorate HFD-induced NAFLD by regulating apoptosis and autophagy through ROS/PI3K/AKT/mTOR signaling pathway. Novel H2S-releasing donors may have therapeutic potential for the treatment of NAFLD.
Collapse
Affiliation(s)
- Dongdong Wu
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China.,School of Stomatology, Henan University, Kaifeng, China
| | - Peiyu Zhong
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Yizhen Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Qianqian Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Jianmei Li
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Zhengguo Liu
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Ailing Ji
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| | - Yanzhang Li
- School of Basic Medical Sciences, Henan University, Kaifeng, China.,Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, China
| |
Collapse
|
33
|
Vaamonde-García C, Burguera EF, Vela-Anero Á, Hermida-Gómez T, Filgueira-Fernández P, Fernández-Rodríguez JA, Meijide-Faílde R, Blanco FJ. Intraarticular Administration Effect of Hydrogen Sulfide on an In Vivo Rat Model of Osteoarthritis. Int J Mol Sci 2020; 21:ijms21197421. [PMID: 33050005 PMCID: PMC7582513 DOI: 10.3390/ijms21197421] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Osteoarthritis (OA) is the most common articular chronic disease. However, its current treatment is limited and mostly symptomatic. Hydrogen sulfide (H2S) is an endogenous gas with recognized physiological activities. The purpose here was to evaluate the effects of the intraarticular administration of a slow-releasing H2S compound (GYY-4137) on an OA experimental model. OA was induced in Wistar rats by the transection of medial collateral ligament and the removal of the medial meniscus of the left joint. The animals were randomized into three groups: non-treated and intraarticularly injected with saline or GYY-4137. Joint destabilization induced articular thickening (≈5% increment), the loss of joint mobility and flexion (≈12-degree angle), and increased levels of pain (≈1.5 points on a scale of 0 to 3). Animals treated with GYY-4137 presented improved motor function of the joint, as well as lower pain levels (≈75% recovery). We also observed that cartilage deterioration was attenuated in the GYY-4137 group (≈30% compared with the saline group). Likewise, these animals showed a reduced presence of pro-inflammatory mediators (cyclooxygenase-2, inducible nitric oxide synthase, and metalloproteinase-13) and lower oxidative damage in the cartilage. The increment of the nuclear factor-erythroid 2-related factor 2 (Nrf-2) levels and Nrf-2-regulated gene expression (≈30%) in the GYY-4137 group seem to be underlying its chondroprotective effects. Our results suggest the beneficial impact of the intraarticular administration of H2S on experimental OA, showing a reduced cartilage destruction and oxidative damage, and supporting the use of slow H2S-producing molecules as a complementary treatment in OA.
Collapse
Affiliation(s)
- Carlos Vaamonde-García
- Grupo de Terapia Celular y Medicina Regenerativa, Universidad de A Coruña, Agrupación Estratégica CICA- INIBIC, Complexo Hospitalario Universitario A Coruña, Campus Oza, 15006 A Coruña, Spain; (C.V.-G.); (Á.V.-A.)
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
| | - Elena F. Burguera
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
- Centro de investigación biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Ángela Vela-Anero
- Grupo de Terapia Celular y Medicina Regenerativa, Universidad de A Coruña, Agrupación Estratégica CICA- INIBIC, Complexo Hospitalario Universitario A Coruña, Campus Oza, 15006 A Coruña, Spain; (C.V.-G.); (Á.V.-A.)
| | - Tamara Hermida-Gómez
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
- Centro de investigación biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Purificación Filgueira-Fernández
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
- Centro de investigación biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Jennifer A. Fernández-Rodríguez
- Grupo de Envejecimiento e Inflamación, Agrupación Estratégica CICA- INIBIC, Complexo Hospitalario Universitario A Coruña, Sergas, Universidad de A Coruña, As Xubias, 15006 A Coruña, Spain;
| | - Rosa Meijide-Faílde
- Grupo de Terapia Celular y Medicina Regenerativa, Universidad de A Coruña, Agrupación Estratégica CICA- INIBIC, Complexo Hospitalario Universitario A Coruña, Campus Oza, 15006 A Coruña, Spain; (C.V.-G.); (Á.V.-A.)
- Correspondence: (R.M.-F.); (F.J.B.); Tel.: +34-981167000 (ext. 5855) (R.M.-F.); +34-981176399 (F.J.B.)
| | - Francisco J. Blanco
- Grupo de Investigación en Reumatología (GIR), INIBIC-Complexo Hospitalario Universitario A Coruña, Sergas, As Xubias, 15006 A Coruña, Spain; (E.F.B.); (T.H.-G.); (P.F.-F.)
- Grupo de Investigación de Reumatología y Salud (GIR), Departamento de Fisioterapia, Medicina y Ciencias Biomédicas, Facultad de Fisioterapia, Agrupación Estrategica CICA-INIBIC, Universidade da Coruña, Campus de Oza, 15006 A Coruña, Spain
- Correspondence: (R.M.-F.); (F.J.B.); Tel.: +34-981167000 (ext. 5855) (R.M.-F.); +34-981176399 (F.J.B.)
| |
Collapse
|
34
|
Han C, Wang Z, Xu Y, Chen S, Han Y, Li L, Wang M, Jin X. Roles of Reactive Oxygen Species in Biological Behaviors of Prostate Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1269624. [PMID: 33062666 PMCID: PMC7538255 DOI: 10.1155/2020/1269624] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Prostate cancer (PCa), known as a heterogenous disease, has a high incidence and mortality rate around the world and seriously threatens public health. As an inevitable by-product of cellular metabolism, reactive oxygen species (ROS) exhibit beneficial effects by regulating signaling cascades and homeostasis. More and more evidence highlights that PCa is closely associated with age, and high levels of ROS are driven through activation of several signaling pathways with age, which facilitate the initiation, development, and progression of PCa. Nevertheless, excessive amounts of ROS result in harmful effects, such as genotoxicity and cell death. On the other hand, PCa cells adaptively upregulate antioxidant genes to detoxify from ROS, suggesting that a subtle balance of intracellular ROS levels is required for cancer cell functions. The current review discusses the generation and biological roles of ROS in PCa and provides new strategies based on the regulation of ROS for the treatment of PCa.
Collapse
Affiliation(s)
- Chenglin Han
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Zilong Wang
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Yingkun Xu
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Shuxiao Chen
- Department of Vascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Yuqing Han
- Department of Radiology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Lin Li
- Department of Orthopedics, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
| | - Muwen Wang
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xunbo Jin
- Department of Urology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250021, China
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| |
Collapse
|
35
|
Wang H, Shi X, Qiu M, Lv S, Zheng H, Niu B, Liu H. Hydrogen Sulfide Plays an Important Role by Influencing NLRP3 inflammasome. Int J Biol Sci 2020; 16:2752-2760. [PMID: 33110394 PMCID: PMC7586428 DOI: 10.7150/ijbs.47595] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 08/12/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammasome is a complex composed of several proteins and an important part of the natural immune system. Nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome is composed of NLRP3, apoptosis associated speck like protein (ASC) and pro-caspase-1. It plays an important role in many diseases. Hydrogen sulfide (H2S) is an important signaling molecule that regulates many physiological and pathological processes. Recent studies indicated that H2S played anti-inflammatory and pro-inflammatory roles in many diseases through influencing NLRP3 inflammasome, but its mechanism was not fully understood. This article reviewed the progress about the effects of H2S on NLRP3 inflammasome and its mechanisms involved in recent years to provide theoretical basis for in-depth study.
Collapse
Affiliation(s)
- Honggang Wang
- Institute of Biomedical Informatics, Bioinformatics Center, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475000, China
| | - Xingzhuo Shi
- School of Life Science, Henan University, Kaifeng, Henan, 475000, China
| | - Mengyuan Qiu
- Institute of Biomedical Informatics, Bioinformatics Center, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475000, China
| | - Shuangyu Lv
- Institute of Biomedical Informatics, Bioinformatics Center, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475000, China
| | - Hong Zheng
- Institute of Biomedical Informatics, Bioinformatics Center, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475000, China
| | - Baohua Niu
- Institute of Biomedical Informatics, Bioinformatics Center, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475000, China
| | - Huiyang Liu
- Institute of Biomedical Informatics, Bioinformatics Center, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475000, China
| |
Collapse
|