1
|
Han B, Tian J, Li J, Chen Y, Liu N, Ma Y, Wang C, Guo X, Liu Y, Zhang Z. Cardioprotective effects of Dendrobium officinale polysaccharides on thiacloprid-induced cardiac injury via modulating mitochondrial dynamics. Int J Biol Macromol 2025; 309:142497. [PMID: 40164262 DOI: 10.1016/j.ijbiomac.2025.142497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/28/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
Thiacloprid (THI), a widely used neonicotinoid pesticide, has been shown to induce cardiac injury, though the underlying mechanisms remain poorly understood. Dendrobium officinale polysaccharides (DOP), a bioactive compound with potent antioxidant properties, may offer protection against such toxicity. This study investigated the cardioprotective effects of DOP in THI-induced cardiac injury in quails, with a particular focus on the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. Network pharmacology analysis identified key targets of DOP, linking them to oxidative stress, mitochondrial dysfunction, and inflammatory pathways. Experimental results demonstrated that DOP significantly reversed THI-induced hematological and biochemical abnormalities, including the restoration of cardiac biomarkers and mitigation of myocardial structural damage. DOP treatment notably activated the Nrf2 pathway, leading to the upregulation of antioxidant enzymes such as heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1), which countered THI-induced oxidative stress. Additionally, DOP restored mitochondrial dynamics by balancing mitochondrial fission and fusion proteins. These findings highlight the central role of Nrf2 activation in the cardioprotective effects of DOP, suggesting that DOP may serve as a promising therapeutic agent for mitigating pesticide-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Biqi Han
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin 150030, China
| | - Jiawen Tian
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin 150030, China
| | - Yuyang Chen
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China
| | - Ning Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China
| | - Yitong Ma
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China
| | - Caihan Wang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China
| | - Xinyu Guo
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China
| | - Yunfeng Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, 150030 Harbin, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin 150030, China.
| |
Collapse
|
2
|
Ebrahimi R, Mohammadpour A, Medoro A, Davinelli S, Saso L, Miroliaei M. Exploring the links between polyphenols, Nrf2, and diabetes: A review. Biomed Pharmacother 2025; 186:118020. [PMID: 40168723 DOI: 10.1016/j.biopha.2025.118020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/03/2025] Open
Abstract
Diabetes mellitus, a complex metabolic disorder, is marked by chronic hyperglycemia that drives oxidative stress and inflammation, leading to complications such as neuropathy, retinopathy, and cardiovascular disease. The Nrf2 pathway, a key regulator of cellular antioxidant defenses, plays a vital role in mitigating oxidative damage and maintaining glucose homeostasis. Dysfunction of Nrf2 has been implicated in the progression of diabetes and its related complications. Polyphenols, a class of plant-derived bioactive compounds, have shown potential in modulating the Nrf2 pathway. Numerous compounds have been found to activate Nrf2 through mechanisms including Keap1 interaction, transcriptional regulation, and epigenetic modification. Preclinical studies indicate their ability to reduce reactive oxygen species (ROS), improve insulin sensitivity, and attenuate inflammation in diabetic models. Clinical trials with certain polyphenols, such as resveratrol, have demonstrated improvements in glycemic parameters, though results remain inconsistent. While polyphenols show promise as a component of non-pharmacological approaches to diabetes management, challenges such as bioavailability, individual variability in response, and limited clinical evidence highlight the need for further investigation. Continued research could enhance understanding of their mechanisms and improve their practical application in mitigating diabetes-related complications.
Collapse
Affiliation(s)
- Reza Ebrahimi
- Faculty of Biological Science and Technology, Department of Cell and Molecular Biology & Microbiology, University of Isfahan, Isfahan, Iran
| | - Alireza Mohammadpour
- Faculty of Biological Science and Technology, Department of Cell and Molecular Biology & Microbiology, University of Isfahan, Isfahan, Iran
| | - Alessandro Medoro
- Department of Medicine and Health Sciences "V.Tiberio", University of Molise, Campobasso 86110, Italy
| | - Sergio Davinelli
- Department of Medicine and Health Sciences "V.Tiberio", University of Molise, Campobasso 86110, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome 00161, Italy.
| | - Mehran Miroliaei
- Faculty of Biological Science and Technology, Department of Cell and Molecular Biology & Microbiology, University of Isfahan, Isfahan, Iran.
| |
Collapse
|
3
|
Froyen EB, Barrantes GP. A Review of the Effects of Flavonoids on NAD(P)H Quinone Oxidoreductase 1 Expression and Activity. J Med Food 2025; 28:407-422. [PMID: 40097203 DOI: 10.1089/jmf.2023.0132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025] Open
Abstract
Cancer is a significant cause of death worldwide. It has been suggested that the consumption of flavonoids decreases the risk for cancer by increasing phase II enzymes, such as Nicotinamide Adenine Dinucleotide Phosphate Hydrogen (NAD(P)H) quinone oxidoreductase 1 (NQO1), glutathione S-transferases, and Uridine 5'-diphospho- (UDP)-glucuronosyltransferases that assist in removing carcinogens from the human body. Flavonoids are bioactive compounds found in a variety of dietary sources, including fruits, vegetables, legumes, nuts, and teas. As such, it is important to investigate which flavonoids are involved in the metabolism of carcinogens to help reduce the risk of cancer. Therefore, the objective of this narrative review was to investigate the effects of commonly consumed flavonoids on NQO1 mRNA expression, protein, and activity in human cell and murine models. PubMed was used to search for peer-reviewed journal articles, which demonstrated that selected flavonoids (e.g., quercetin, apigenin, luteolin, genistein, and daidzein) increase NQO1, and therefore, increase the excretion of carcinogens. However, more research is needed regarding the mechanisms by which flavonoids induce NQO1. Furthermore, it is suggested that future efforts focus on providing precise flavonoid recommendations to decrease the risk factors for chronic diseases.
Collapse
Affiliation(s)
- Erik B Froyen
- Department of Nutrition and Food Science, Huntley College of Agriculture, California State Polytechnic University, Pomona, California, USA
| | - Gianluis Pimentel Barrantes
- Department of Nutrition and Food Science, Huntley College of Agriculture, California State Polytechnic University, Pomona, California, USA
| |
Collapse
|
4
|
Hayakawa R, Ishii T, Fushimi T, Kamei Y, Yamaguchi A, Sugimoto K, Ashida H, Akagawa M. Luteolin protects human ARPE-19 retinal pigment epithelium cells from blue light-induced phototoxicity through activation of Nrf2/Keap1 signaling. Free Radic Res 2025; 59:356-368. [PMID: 40340707 DOI: 10.1080/10715762.2025.2503832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/08/2025] [Accepted: 04/23/2025] [Indexed: 05/10/2025]
Abstract
Age-related macular degeneration (AMD), a serious physical and mental health problem worldwide, is the leading cause of irreversible, severe vision impairment and loss in older people. AMD is associated with multiple risk factors, many of which are closely linked to increased oxidative stress. Some studies have suggested that long-term and excessive exposure to blue light may be a potential risk factor for the development or progression of AMD. Recently, we demonstrated that blue light irradiation caused oxidative stress in all-trans-retinal (atRAL)-exposed human ARPE-19 retinal pigment epithelium cells by generating singlet oxygen (1O2), leading to apoptotic cell death. Luteolin, a flavonoid found in various edible plants, has been reported to possess divergent health-promoting properties including anti-oxidative and chemopreventive effects by up-regulating anti-oxidative and phase II detoxifying enzymes through activation of Keap1/Nrf2 signaling. Herein, we verified the cytoprotective action of luteolin against blue light irradiation using atRAL-exposed ARPE-19 cells. Our results established that luteolin effectively prevented blue light-induced apoptosis of ARPE-19 cells by mitigating oxidative stress. We also confirmed that luteolin suppressed intracellular accumulation of 1O2 and formation of atRAL-derived lipofuscin by increased expression of heme oxygenase-1 and aldehyde dehydrogenase 1A1 through activation of Keap1/Nrf2 signaling. Furthermore, our data implied that the luteolin-provoked activation of Keap1/Nrf2 signaling might be due to covalent binding of luteolin o-quinone to the critical cysteinyl thiol in Keap1. The present results suggest that luteolin could be helpful in the prevention and amelioration of blue light-induced retinal degeneration, including AMD.
Collapse
Affiliation(s)
- Ryo Hayakawa
- Department of Biological Chemistry, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Japan
| | - Takeshi Ishii
- Faculty of Nutrition, Kobe Gakuin University, Kobe, Japan
| | - Taiki Fushimi
- Department of Food and Nutrition, Institute of Biomedical sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuki Kamei
- Department of Food and Nutrition, Institute of Biomedical sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Ai Yamaguchi
- Live Cell Imaging Institute, Osaka, Japan
- Research Institute for Cellular Damage Evaluation of Ionizing Radiation and Blue Light, Osaka Prefecture University, Sakai, Japan
| | - Kenji Sugimoto
- Live Cell Imaging Institute, Osaka, Japan
- Research Institute for Cellular Damage Evaluation of Ionizing Radiation and Blue Light, Osaka Prefecture University, Sakai, Japan
| | - Hitoshi Ashida
- Faculty of Food and Nutrition, Mukogawa Women's University, Nishinomiya, Japan
| | - Mitsugu Akagawa
- Department of Food and Nutrition, Institute of Biomedical sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
5
|
Farhadi R, Daniali M, Baeeri M, Khorasani R, Haghi-Aminjan H, Gholami M, Rahimifard M, Navaei-Nigjeh M, Abdollahi M. Molecular evidence of the inhibitory potential of melatonin against sodium arsenite toxicity. Heliyon 2025; 11:e42113. [PMID: 39916822 PMCID: PMC11799970 DOI: 10.1016/j.heliyon.2025.e42113] [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: 07/10/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
Introduction Sodium arsenite (SA), NaAsO2, is among the most hazardous toxicants, and wide use and presence of this toxicant leads to a severe environmental threat. Exposure to SA is associated with many health concerns, such as the prevalence of cancer and diabetes mellitus type 2 (DMT2). Many studies suggest that SA induces inflammation and biochemical impairments through different mechanisms, including increasing oxidative stress and altering vital genes such as biochemical and anti-inflammatory. Recent studies on melatonin (MLT), a harmless hormone secreted in the body generally for induction of sleepiness, find many beneficial and positive effects. Mitigating different harms and toxicities through different mechanisms, such as antioxidant properties, anti-inflammatory effects, and critical gene regulation, is essential. Due to these findings, this study aimed to evaluate the hypothesis that MLT may ameliorate pancreatic damage caused by exposure to SA. Methods Forty-eight adult healthy male wistar rats aged 7-8 weeks were divided into eight for this research. Group 1 did not receive any intervention. Group 2 received 10 mg/kg/day MLT through intraperitoneal (IP) injection. Groups 3, 4, and 5 received 1.5 (1/10 LD50), 5 (1/3 LD50), and 7.5 (1/2 LD50) mg/kg SA, respectively. Groups 6, 7, and 8 were given 1.5 (1/10 LD50), 5 (1/3 LD50), and 7.5 (1/2 LD50) mg/kg of SA along with 10 mg/kg/day MLT, respectively, during the last ten days of the experiment. After 28 days of the experiment, the blood and tissue samples of the pancreas were removed for biochemical and pathological examination. Results MLT attenuates SA toxicity by reducing oxidative stress biomarkers and inflammation markers. Moreover, MLT improves SA exposure's biochemical and functional damages by regulating related genes and pathways. Conclusion MLT poses protective and preventive effects on the pancreas against exposure to SA. However, MLT's therapeutic and beneficial impacts have great potential for further investigation.
Collapse
Affiliation(s)
- Ramtin Farhadi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Marzieh Daniali
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Baeeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Reza Khorasani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamed Haghi-Aminjan
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mahban Rahimifard
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mona Navaei-Nigjeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Toxicology and Diseases Specialty Group, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
6
|
Zhao R, Xu S, Jia C, Zhu S, Ma L, Chen Y, Chen D. Exploring the protective role of Heracleum persicum L. extract in testicular toxicity induced by mercuric chloride: insights into hormonal modulation and cell survival pathways. Toxicol Res (Camb) 2025; 14:tfaf015. [PMID: 39906185 PMCID: PMC11788597 DOI: 10.1093/toxres/tfaf015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/12/2025] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
The study investigated the effects of Heracleum persicum L. extract (HPE) on oxidative damage caused by mercuric chloride (HgCl₂) in rat testes. Sixty male Wistar rats were divided into six groups: a sham group, a HgCl₂ group, three groups receiving HgCl₂ with HPE at doses of 250, 500, and 750 mg/kg, and a control group treated with 750 mg/kg HPE alone over 50 days. HgCl₂ was administered intraperitoneally for the first 10 days, followed by HPE gavage for 40 days. On day 51, hormone levels (testosterone, FSH, LH), nitric oxide levels, antioxidant enzyme activity, and pro-inflammatory cytokines were measured. Testicular tissue was analyzed for thiobarbituric acid reactive substances, ferric reducing capacity, thiol levels, and stereological indicators of seminiferous tubules. The study also examined the p53/Cas-3/Bax/Bcl-2 apoptotic pathway. LC-ESI/MS and SEM-EDS analysis detected 25 substances and 14 mineral elements. HgCl₂ exposure significantly reduced LH, T, and FSH levels, while HPE improved these hormones, especially at higher doses. Inflammatory cytokines were elevated due to HgCl₂, but HPE reduced (P < 0.05) these levels and enhanced (P < 0.05) antioxidant enzyme activity, indicating protective effects against oxidative stress. Testicular analysis showed significant (P < 0.05) damage from HgCl₂, but HPE preserved tissue integrity and improved parameters. Weight measurements indicated that HgCl₂ reduced (P < 0.05) body and reproductive weights, while HPE restored these weights. HPE also counteracted apoptotic changes, highlighting its potential as a therapeutic agent against HgCl₂-induced damage.
Collapse
Affiliation(s)
- Ronghui Zhao
- Department of Clinical Pharmacy Office of Pharmacy, School of Clinical Medical, Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi district, Baoding, China
- Department of Pharmacy, School of Clinical Medical, Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi district, Baoding, China
- College of Chemistry and Materials Science, Hebei University, No. 180 Wusi Dong Road, Lian Chi District, Baoding, China
| | - Shijuan Xu
- Department of Library of Scientific Research, School of Clinical Medical, Affiliated Hospital of Hebei University, No. 180 Wusi Dong Road, Lian Chi District, Baoding, China
| | - Chao Jia
- Department of Clinical Pharmacy Office of Pharmacy, School of Clinical Medical, Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi district, Baoding, China
| | - Shufang Zhu
- Department of Clinical Pharmacy Office of Pharmacy, School of Clinical Medical, Affiliated Hospital of Hebei University, No. 212 Yuhua East Road, Lianchi district, Baoding, China
| | - Lianshun Ma
- Morphological laboratory, School of Basic Medical Sciences, Hebei University, Baoding, China
| | - Yalan Chen
- Department of Gastroenterology, Affiliated Hospital of Hebei University, School of Clinical Medical, No. 212 Yuhua East Road, Lianchi district, Baoding, China
| | - Dalei Chen
- Department of Gastroenterology, Affiliated Hospital of Hebei University, School of Clinical Medical, No. 212 Yuhua East Road, Lianchi district, Baoding, China
| |
Collapse
|
7
|
Ilhan I, Asci H, Candan IA, Savran M, Imeci OB, Sevuk MA. Cannabidiol mitigates methotrexate-induced hepatic injury via SIRT-1/p53 signaling and mitochondrial pathways: reduces oxidative stress and inflammation. Drug Chem Toxicol 2025; 48:210-218. [PMID: 39603835 DOI: 10.1080/01480545.2024.2425994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 10/22/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024]
Abstract
Methotrexate (MTX), a widely used chemotherapeutic agent, often induces hepatotoxicity, limiting its clinical utility. Cannabidiol (CBD), derived from hemp, possesses antioxidant, anti-inflammatory, and antiapoptotic properties. This study aims to investigate CBD's protective effects against MTX-induced liver injury and elucidate the underlying mechanisms. Thirty-two female Wistar Albino rats were divided into four groups: control, MTX (20 mg/kg intraperitoneally [i.p.] once), MTX+CBD (20 mg/kg i.p. once + 5 mg/kg i.p. for seven days), and CBD (5 mg/kg, i.p. for seven days). Biochemical analyses of serum and liver tissues were performed to assess oxidative stress markers (total oxidant status, total antioxidant status, oxidative stress index), liver function tests (AST, ALT), and antioxidant enzyme activities (glutathione peroxidase, superoxide dismutase). Histopathological and immunohistochemical examinations were conducted to evaluate liver tissue damage and TNF-α expression. Genetic analyses were performed to measure the expression levels of SIRT-1, p53, Bcl-2, and Bax genes using RT-qPCR. MTX administration increased oxidative stress markers, liver enzymes, TNF-α, p53, and Bax levels while decreasing antioxidant defenses and SIRT-1 expression. CBD administration reversed these alterations effectively. CBD mitigated MTX-induced hepatotoxicity by reducing oxidative stress, inflammation, and apoptosis. It activates antioxidant defenses via SIRT-1 upregulation, suppresses inflammation by reducing TNF-α, and prevents apoptosis by modulating p53, Bcl-2, and Bax gene expressions. These findings suggest CBD could be a promising therapeutic agent for chemotherapy-induced liver damage. Further research is warranted to explore additional pathways and broader molecular mechanisms.
Collapse
Affiliation(s)
- Ilter Ilhan
- Department of Biochemistry, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Halil Asci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Ibrahim Aydın Candan
- Department of Histology and Embryology, Alanya Alaaddin Keykubat University, Antalya, Turkey
| | - Mehtap Savran
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Orhan Berk Imeci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Mehmet Abdulkadir Sevuk
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
8
|
Li J, Guo C, Liu Y, Han B, Lv Z, Jiang H, Li S, Zhang Z. Chronic arsenic exposure-provoked biotoxicity involved in liver-microbiota-gut axis disruption in chickens based on multi-omics technologies. J Adv Res 2025; 67:373-386. [PMID: 38237767 PMCID: PMC11725159 DOI: 10.1016/j.jare.2024.01.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 10/27/2023] [Accepted: 01/13/2024] [Indexed: 01/25/2024] Open
Abstract
INTRODUCTION Arsenic has been ranked as the most hazardous substance by the U.S. Agency for Toxic Substances and Disease Registry. Environmental arsenic exposure-evoked health risks have become a vital public health concern worldwide owing to the widespread existence of arsenic. Multi-omics is a revolutionary technique to data analysis providing an integrated view of bioinformation for comprehensively and systematically understanding the elaborate mechanism of diseases. OBJECTIVES This study aimed at uncovering the potential contribution of liver-microbiota-gut axis in chronic inorganic arsenic exposure-triggered biotoxicity in chickens based on multi-omics technologies. METHODS Forty Hy-Line W-80 laying hens were chronically exposed to sodium arsenite with a dose-dependent manner (administered with drinking water containing 10, 20, or 30 mg/L arsenic, respectively) for 42 d, followed by transcriptomics, serum non-targeted metabolome, and 16S ribosomal RNA gene sequencing accordingly. RESULTS Arsenic intervention induced a serious of chicken liver dysfunction, especially severe liver fibrosis, simultaneously altered ileal microbiota populations, impaired chicken intestinal barrier, further drove enterogenous lipopolysaccharides translocation via portal vein circulation aggravating liver damage. Furtherly, the injured liver disturbed bile acids (BAs) homoeostasis through strongly up-regulating the BAs synthesis key rate-limiting enzyme CYP7A1, inducing excessive serum total BAs accumulation, accompanied by the massive synthesis of primary BA-chenodeoxycholic acid. Moreover, the concentrations of secondary BAs-ursodeoxycholic acid and lithocholic acid were markedly repressed, which might involve in the repressed dehydroxylation of Ruminococcaceae and Lachnospiraceae families. Abnormal BAs metabolism in turn promoted intestinal injury, ultimately perpetuating pernicious circle in chickens. Notably, obvious depletion in the abundance of four profitable microbiota, Christensenellaceae, Ruminococcaceae, Muribaculaceae, and Faecalibacterium, were correlated tightly with this hepato-intestinal circulation process in chickens exposed to arsenic. CONCLUSION Our study demonstrates that chronic inorganic arsenic exposure evokes liver-microbiota-gut axis disruption in chickens and establishes a scientific basis for evaluating health risk induced by environmental pollutant arsenic.
Collapse
Affiliation(s)
- Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Changming Guo
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yan Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Biqi Han
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Zhanjun Lv
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Huijie Jiang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China.
| |
Collapse
|
9
|
Mahwish, Imran M, Naeem H, Hussain M, Alsagaby SA, Al Abdulmonem W, Mujtaba A, Abdelgawad MA, Ghoneim MM, El‐Ghorab AH, Selim S, Al Jaouni SK, Mostafa EM, Yehuala TF. Antioxidative and Anticancer Potential of Luteolin: A Comprehensive Approach Against Wide Range of Human Malignancies. Food Sci Nutr 2025; 13:e4682. [PMID: 39830909 PMCID: PMC11742186 DOI: 10.1002/fsn3.4682] [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: 08/01/2024] [Revised: 10/25/2024] [Accepted: 11/30/2024] [Indexed: 01/22/2025] Open
Abstract
Luteolin is widely distributed phytochemical, a flavonoid, in kingdom plantae. Luteolin with potential antioxidant activity prevent ROS-induced damages and reduce oxidative stress which is mainly responsible in pathogenesis of many diseases. Several chemo preventive activities and therapeutic benefits are associated with luteolin. Luteolin prevents cancer via modulation of numerous pathways, that is, by inactivating proteins; such as procaspase-9, CDC2 and cyclin B or upregulation of caspase-9 and caspase-3, cytochrome C, cyclin A, CDK2, and APAF-1, in turn inducing cell cycle arrest as well as apoptosis. It also enhances phosphorylation of p53 and expression level of p53-targeted downstream gene. By Increasing BAX protein expression; decreasing VEGF and Bcl-2 expression it can initiate cell cycle arrest and apoptosis. Luteolin can stimulate mitochondrial-modulated functions to cause cellular death. It can also reduce expression levels of p-Akt, p-EGFR, p-Erk1/2, and p-STAT3. Luteolin plays positive role against cardiovascular disorders by improving cardiac function, decreasing the release of inflammatory cytokines and cardiac enzymes, prevention of cardiac fibrosis and hypertrophy; enhances level of CTGF, TGFβ1, ANP, Nox2, Nox4 gene expressions. Meanwhile suppresses TGFβ1 expression and phosphorylation of JNK. Luteolin helps fight diabetes via inhibition of alpha-glucosidase and ChE activity. It can reduce activity levels of catalase, superoxide dismutase, and GS4. It can improve blood glucose, insulin, HOMA-IR, and HbA1c levels. This review is an attempt to elaborate molecular targets of luteolin and its role in modulating irregularities in cellular pathways to overcome severe outcomes during diseases including cancer, cardiovascular disorders, diabetes, obesity, inflammation, Alzheimer's disease, Parkinson's disease, hepatic disorders, renal disorders, brain injury, and asthma. As luteolin has enormous therapeutic benefits, it could be a potential candidate in future drug development strategies.
Collapse
Affiliation(s)
- Mahwish
- Institute of Food Science and NutritionUniversity of SargodhaSargodhaPakistan
| | - Muhammad Imran
- Department of Food Science and TechnologyUniversity of NarowalNarowalPakistan
| | - Hammad Naeem
- Department of Food Science and TechnologyMuhammad Nawaz Shareef University of AgricultureMultanPakistan
| | - Muzzamal Hussain
- Department of Food SciencesGovernment College University FaisalabadFaisalabadPakistan
| | - Suliman A. Alsagaby
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesMajmaah UniversityAL‐MajmaahSaudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of MedicineQassim UniversityBuraidahSaudi Arabia
| | - Ahmed Mujtaba
- Department of Food Sciences and Technology, Faculty of Engineering and TechnologyHamdard University Islamabad campusIslamabadPakistan
| | - Mohamed A. Abdelgawad
- Department of Pharmaceutical Chemistry, College of PharmacyJouf UniversityAljoufSaudi Arabia
| | - Mohammed M. Ghoneim
- Department of Pharmacy Practice, College of PharmacyAlMaarefa UniversityRiyadhSaudi Arabia
| | - Ahmed H. El‐Ghorab
- Department of Chemistry, College of ScienceJouf UniversitySakakaSaudi Arabia
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical SciencesJouf UniversitySakakaSaudi Arabia
| | - Soad K. Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of MedicineKing Abdulaziz UniversityJeddahSaudi Arabia
| | - Ehab M. Mostafa
- Department of Pharmacognosy, College of PharmacyJouf UniversitySakakaSaudi Arabia
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys)Al‐Azhar UniversityCairoEgypt
| | - Tadesse Fenta Yehuala
- Faculty of Chemical and Food Engineering, Bahir Dar Institute of TechnologyBahir Dar UniversityBahir DarEthiopia
| |
Collapse
|
10
|
Zhu X, Hai Z, Ning Z. Salidroside impedes Ang II-infused myocardial fibrosis by activating the SIRT1-Nrf2 pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2025; 28:815-824. [PMID: 40343296 PMCID: PMC12057753 DOI: 10.22038/ijbms.2025.83659.18105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 01/01/2025] [Indexed: 05/11/2025]
Abstract
Objectives This research examined the protective function of salidroside (SAL) against angiotensin II (Ang II)-infused myocardial fibrosis and its associated mechanism. Materials and Methods The C57BL/6 male murine models (n=24) received either saline solution or Ang II (1500 ng/kg/day) subcutaneously and an oral dosage of SAL (50 mg/kg/day) once daily for 28 days. Newborn Sprague-Dawley (SD) rats were used to isolate atrial fibroblasts. Results The fibrotic region was raised by Ang II infusion, while SAL treatment inhibited it. Collagen I and III expression was raised by Ang II induction, but SAL therapy reduced their expression. SAL therapy also decreased the expression of other fibroblast differentiation-related markers induced by Ang II infusion. It elevated SIRT1, Nrf2, and HO-1 levels in atrial fibroblasts. Additionally, SAL significantly inhibited atrial fibroblasts, whereas EX527, an inhibitor of SIRT1, noticeably increased the migration ability. Furthermore, SAL suppressed intracellular ROS production and oxidative stress in Ang II-infused atrial fibroblasts. Conclusion SAL protects against myocardial fibrosis infused by Ang II by activating the SIRT1-Nrf2 pathway.
Collapse
Affiliation(s)
| | | | - Zhongping Ning
- Department of Cardiology, Shanghai Pudong New Area Zhoupu Hospital (Shanghai Health Medical College Affiliated Zhoupu Hospital) , Shanghai 201318, China
| |
Collapse
|
11
|
Stephen Adeyemi O, Emmanuel Rotimi D, Demilade Fatinukun H, Oluwadamilare Adeogun V, Owen Evbuomwan I, Lateef Adebayo O, Atolani O, Adewumi Akanji M. Antioxidant and inflammatory-modulating properties of ginger and bitterleaf teas. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:4122-4136. [PMID: 38591815 DOI: 10.1080/09603123.2024.2338894] [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: 11/28/2023] [Accepted: 03/30/2024] [Indexed: 04/10/2024]
Abstract
The present study evaluated the effects of ginger and bitterleaf tea infusions on redox and inflammatory balance in rats. Twenty-four Wistar rats with weights of between 160 and 180 g were assigned into four (4) groups (n = 6). The control group received distilled water, while the remaining groups were administered tea infusions of ginger, bitterleaf, or a combination of both at 5 mg/mL, respectively. Bitterleaf and ginger teas elevated the levels of superoxide dismutase, catalase, glutathione peroxidase, and reduced glutathione in rat plasma and liver, while malondialdehyde levels decreased. Furthermore, ginger tea caused an increase in the expression of nuclear factor erythroid-2-related factor 2 (Nrf-2) and reduced tumor necrosis factor alpha (TNF-α). The GC-MS analysis of the teas identified 77 chemical compounds, among which gingerol and precocene I were predominant. Collectively, the findings indicate, in particular, that ginger tea may boost antioxidant and anti-inflammatory capacity by increasing Nrf-2 levels.
Collapse
Affiliation(s)
- Oluyomi Stephen Adeyemi
- Department of Biochemistry, Medicinal Biochemistry, Nanomedicine & Toxicology Laboratory, Bowen University, Iwo, Nigeria
- Laboratory of Sustainable Animal Environment, Graduate School of Agricultural Science, Tohoku University, Osaki, Miyagi, Japan
| | - Damilare Emmanuel Rotimi
- SDG 03 Group - Good Health & Well-being, Landmark University, Omu-Aran, Kwara State, Nigeria
- Department of Biochemistry, Medicinal Biochemistry, Nanomedicine & Toxicology Laboratory, Landmark University, Omu-Aran, Nigeria
| | - Heritage Demilade Fatinukun
- SDG 03 Group - Good Health & Well-being, Landmark University, Omu-Aran, Kwara State, Nigeria
- Department of Biochemistry, Medicinal Biochemistry, Nanomedicine & Toxicology Laboratory, Landmark University, Omu-Aran, Nigeria
| | - Victor Oluwadamilare Adeogun
- SDG 03 Group - Good Health & Well-being, Landmark University, Omu-Aran, Kwara State, Nigeria
- Department of Biochemistry, Medicinal Biochemistry, Nanomedicine & Toxicology Laboratory, Landmark University, Omu-Aran, Nigeria
| | - Ikponmwosa Owen Evbuomwan
- Department of Microbiology, Cellular Parasitology Unit, College of Pure and Applied Sciences, Landmark University, Omu-Aran, Nigeria
| | - Olusegun Lateef Adebayo
- Department of Biochemistry, Faculty of Basic Medical Sciences, Redeemer's University, Osun State, Nigeria
| | | | | |
Collapse
|
12
|
Abdelzaher WY, Geddawy A, Attya ME, Ali AHSA, Elroby Ali DM, Waggas DS, Alshaeri HK, Ibrahim YF. Sirt1/Nrf2/TNFα; TLR4/Myd88/NF-κB signaling pathways are involved in mediating hepatoprotective effect of bupropion in rat model of myocardial infarction. Immunopharmacol Immunotoxicol 2024; 46:872-883. [PMID: 39390633 DOI: 10.1080/08923973.2024.2415461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND The aim of the current study is to identify the possible protective effect of bupropion (BUP) on liver injury in rat model of myocardial infarction (MI). BUP was administered in the presence and absence of MI. MATERIALS AND METHODS Thirty-two Wistar adult male rats were randomly arranged into four groups: control, BUP (30 mg/kg/day, intraperitoneal) for 28 days, isoproterenol (ISO) was injected subcutaneous (85 mg/kg) in the 26th and 27th days and BUP/ISO groups. Cardiac and hepatic enzymes were measured, also Hepatic oxidative stress indicators, as well as inflammatory and apoptotic biomarkers, were evaluated. Cardiac and hepatic histopathological examination and hepatic nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) immunohistochemical study were also detected. RESULTS ISO significantly increased cardiac and hepatic enzymes, hepatic oxidative stress, inflammatory, apoptotic, with a histopathological picture of cardiac and hepatic damage and high hepatic NF-κB immunoexpression were detected. BUP significantly normalized the upraised oxidative, inflammatory, and apoptotic parameters, with an impressive improvement in the histopathological picture and a reduction in hepatic NF-κB immunoexpression. CONCLUSION BUP protects against liver injury on top of MI in rat model via modulation of Sirtuin type 1 (Sirt1)/Nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/tumor necrosis factor α (TNFα); Toll-like receptor 4 (TLR4)/Hepatic myeloid differentiation primary response 88(Myd88)/NF-κB signaling pathways.
Collapse
Affiliation(s)
| | - Ayman Geddawy
- Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al Kharj, Saudia Arabia
| | - Mina Ezzat Attya
- Department of Pathology, Faculty of Medicine, Minia University, Minia, Egypt
| | | | - Doaa Mohamed Elroby Ali
- Department of Biochemistry and molecular biology, Faculty of Pharmacy, Sohag University, Sohag, Egypt
| | - Dania S Waggas
- Pathological Sciences Department- MBBS Program, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Heba K Alshaeri
- Pharmaceutical Sciences Department- PharmD Program, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | - Yasmine F Ibrahim
- Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
13
|
Mumtaz F, Farag BM, Farahat MA, Farouk FA, Aarif MY, Eltresy MH, Amin MH, Habotta OA, Alneghery LM, Alawam AS, Almuqri EA, Aleissa MS, Alhudhaibi AM, Al-Olayan E, Abdel Moneim AE, Ramadan SS. Leek (Allium ampeloprasum var. kurrat) aqueous extract loaded on selenium nanoparticles protects against testis and brain injury induced by mercuric chloride in rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:9062-9075. [PMID: 38993070 DOI: 10.1002/jsfa.13733] [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: 02/13/2024] [Revised: 05/11/2024] [Accepted: 06/24/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Mercuric chloride (HgCl2) is poisonous to humans and animals and typically damages the nervous system and other organs. Mercuric chloride exposition disclosed to initiation of oxidative stress pathway can result in a defect in male fertility and testis tissue. Synthesized selenium nanoparticles (SeNPs) were characterized with a diameter range minimal than 100 nm, having the effective sets of the biological matter. The present study aimed to evaluate the effect of biosynthesized SeNPs, prepared by leek extract on Wistar rats' testicles and brain. METHODS Thirty-five Wistar male rats (120-150 g) were randomly split into five groups (n = 7), orally ingested with leek aqueous extract loaded on SeNPs, and then the animals were administered with mercury II chloride (HgCl2) to induce testis injury and damage the nervous system. RESULTS The used dose of mercuric chloride led to oxidative stress damage in the testis of the rats which was evidenced by a decrease in testosterone, luteinizing hormone (LH), follicle-stimulating hormone (FSH) and proliferating cell nuclear antigen (PCNA) levels, and an increase in nuclear factor-kappa B (NF-κB) and caspase-3. Also, HgCl2 decreased the levels of dopamine (DA), serotonin (5-HT), norepinephrine (NE) and brain-derived neurotrophic factor (BDNF) in the brains of rats. In addition, A decrease was observed in the levels of antioxidant markers, B-cell lymphoma-2 (Bcl-2), as well as an increase in malondialdehyde (MDA), nitric oxide (NO), NF-κB, tumor necrosis factor (TNF)-α, interleukin (IL)-1β and Bax in both testes and brains. Pre-treatment with leek extract loaded on SeNPs significantly ameliorated testosterone, LH, FSH, PCNA and caspase-3 levels in the testis and DA, 5-HT, NE and BDNF in brains. Although the contents of MDA, NO, TNF-α, IL-1β, NF-κB and Bax decreased significantly in both. glutathione, glutathione peroxidase, glutathione reductase, catalase, superoxide dismutase and Bcl-2 levels were significantly improved in both organs. CONCLUSION Our findings suggest that treatment with aqueous leek extract loaded on SeNPs may offer promising prospects for the advancement of anti-inflammation activity against testis injury and also have a very key role in neurobehavioral alterations as a result of mercury toxicity. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Farah Mumtaz
- Department of Biology, Collage of Science, University of Babylon, Babylon, Iraq
| | - Bahaa M Farag
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Mennatullah A Farahat
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Fatma A Farouk
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Moataz Y Aarif
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Mohamed H Eltresy
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Menna H Amin
- Biochemistry Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Lina M Alneghery
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Abdullah S Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Eman A Almuqri
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Mohammed S Aleissa
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Abdulrahman M Alhudhaibi
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, Egypt
| | - Shimaa S Ramadan
- Biochemistry Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
14
|
Yu H, Huang L, Gui L, Wu Z, Luo H, Xu M, Zhang Y, Qian Y, Cao W, Liu L, Li F. Luteolin ameliorates hyperuricemic nephropathy by activating urate excretion and Nrf2/HO-1/NQO1 antioxidant pathways in mice. Food Sci Nutr 2024; 12:8053-8066. [PMID: 39479625 PMCID: PMC11521689 DOI: 10.1002/fsn3.4403] [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: 06/02/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 11/02/2024] Open
Abstract
Luteolin is a natural flavonoid, which exists in many plants, including onions, broccoli, carrots, peppers, celery, olive oil, and mint. Luteolin is a dietary flavonoid with potent uric acid-lowering and antioxidant bioactivities. To date, the mechanism by which luteolin alleviates hyperuricemia nephropathy (HN) still needs to be better defined. This study aims to evaluate the therapeutic efficacy of luteolin in a preclinical mouse model and in vitro. Luteolin was administered in the HN mice induced by the combination of potassium oxonate and hypoxanthine to evaluate the potential renoprotective effects in vivo. The NRK-52E cells were stimulated with adenosine for in vitro evaluation. Hematoxylin and eosin staining, biochemical analysis, immunoblotting, immunofluorescence, and immunohistochemistry were performed for the histopathologic and mechanistic investigations. The results suggest that luteolin attenuated tubular dilation and epithelial atrophy in the renal tissue of HN mice. Further, luteolin improved biochemical indicators concerning renal functions and oxidative stress in vivo. Mechanistically, luteolin reduced the renal expressions of KIM-1 and caspase-3. Luteolin activated renal SIRT1/6 cascade and its downstream Nrf2-mediated antioxidant pathway. Furthermore, luteolin elevated the renal expressions of ATP-binding cassette subfamily G isoform 2 protein (ABCG2) and organic anion/cation transporters. In addition, livers of luteolin-treated HN mice exhibited robust inhibition of xanthine oxidase. Together, our study shows that luteolin alleviates renal injury in the HN mice by activating urate excretion and Nrf2/HO-1/NQO1 antioxidant pathways and inhibiting liver xanthine oxidase activity. Thus, luteolin may be a potential agent for the treatment of HN.
Collapse
Affiliation(s)
- Huifan Yu
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
- Institute of BiomedicineHubei University of MedicineShiyanHubeiChina
| | - Linsheng Huang
- Department of Hepatopancreatobiliary Surgery, Taihe HospitalHubei University of MedicineShiyanHubeiChina
| | - Lili Gui
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
- Institute of BiomedicineHubei University of MedicineShiyanHubeiChina
| | - Zhengkun Wu
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
- Institute of BiomedicineHubei University of MedicineShiyanHubeiChina
| | - Han Luo
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
| | - Mao Xu
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
| | - Yan Zhang
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
| | - Yongshuai Qian
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
| | - Wenjie Cao
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
| | - Li Liu
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
| | - Fei Li
- School of Pharmaceutical Sciences, Hubei Key Laboratory of Wudang Local Chinese Medicine ResearchHubei University of MedicineShiyanHubeiChina
| |
Collapse
|
15
|
Sedky A, Famurewa AC. Anti-ischemic drug trimetazidine blocks mercury nephrotoxicity by suppressing renal redox imbalance, inflammatory stress and caspase-dependent apoptosis in rats. Drug Chem Toxicol 2024; 47:674-681. [PMID: 37528808 DOI: 10.1080/01480545.2023.2242007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/12/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023]
Abstract
Trimetazidine (TMZ) is a promising emerging therapeutic piperazine derivative for renal pathologies. However, the nephroprotective mechanism of TMZ against heavy metal-induced toxicity is unknown. This study, therefore, aimed to explore whether TMZ could mitigate mercury-induced nephrotoxicity in rats. Rats were injected TMZ (3 mg/kg bw) and/or mercury chloride (HgCl2) (4 mg/kg bw) for 4 days (n = 6 rats per group). The blood analysis revealed marked increases in creatinine, urea and uric acid levels in HgCl2 group compared to the control. HgCl2 induced prominent decreases in renal superoxide dismutase (SOD), catalase (CAT), glutathione peroxide (GPx) activities compared to the control followed by marked increases in the levels of malondialdehyde (MDA), interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), caspase-3 and caspase-9. Whereas the renal levels of anti-inflammatory cytokines interleukin-4 (IL-4) and interleukin-10 (IL-10) reduced considerably compared to the control. Contrarily, it was found that in the rats administered TMZ + HgCl2, levels of renal markers, MDA, TNF-α, IL-6 and caspases-3/-9 were prominently reduced compared to the HgCl2 group. The renal SOD, CAT, GPx, IL-4, and IL-10 were markedly elevated along with ameliorated histopathological lesions. On the whole, therefore, TMZ could be repurposed for blocking HgCl2 nephrotoxicity via inhibition of oxidative inflammation and apoptosis in rats.
Collapse
Affiliation(s)
- Azza Sedky
- Department of Biological Sciences, College of Science, King Faisal University, Saudi Arabia
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo, Nigeria
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
16
|
Shyam M, Sabina EP. Harnessing the power of Arctium lappa root: a review of its pharmacological properties and therapeutic applications. NATURAL PRODUCTS AND BIOPROSPECTING 2024; 14:49. [PMID: 39162715 PMCID: PMC11335715 DOI: 10.1007/s13659-024-00466-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024]
Abstract
Arctium lappa, widely recognized as burdock, is a perennial plant that is employed in the realm of traditional Chinese medicine for a wide range of medicinal applications. The herb is rich in bioactive metabolites with therapeutic potential, encompassing polyphenolic antioxidants in its leaves, and flavonoids and fructo-oligosaccharides in its underground parts. Nutraceuticals originating from botanical sources such as Arctium lappa provide supplementary health advantages alongside their nutritional content and have demonstrated effectiveness in the prevention and management of specific ailments. The utilization of Arctium lappa root extract has exhibited encouraging outcomes in addressing hepatotoxicity induced by cadmium, lead, chromium, and acetaminophen, ameliorating liver damage and oxidative stress. Additionally, the root extract displays properties such as antidiabetic, hypolipidemic, aphrodisiac, anti-rheumatic, anti-Alzheimer, and various other pharmacological actions.
Collapse
Affiliation(s)
- Mukul Shyam
- Department of Biotechnology, School of Biosciences and Technology, VIT University, SBST, VIT, Vellore, 632014, Tamil Nadu, India
| | - Evan Prince Sabina
- Department of Biotechnology, School of Biosciences and Technology, VIT University, SBST, VIT, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
17
|
Rahman AA, Hegazy A, Elabbasy LM, Shoaeir MZ, Abdel-Aziz TM, Abbas AS, Khella HWZ, Eltrawy AH, Alshaman R, Aloyouni SY, Aldahish AA, Zaitone SA. Leflunomide-induced cardiac injury in adult male mice and bioinformatic approach identifying Nrf2/NF-κb signaling interplay. Toxicol Mech Methods 2024; 34:639-653. [PMID: 38389224 DOI: 10.1080/15376516.2024.2322666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Leflunomide (LFND) is an immunosuppressive and immunomodulatory disease-modifying antirheumatic drug (DMARD) that was approved for treating rheumatoid arthritis. LFND-induced cardiotoxicity was not fully investigated since its approval. We investigated the cardiac injury in male mice and identified the role of nuclear factor erythroid 2-related factor 2/nuclear factor-κ B (Nrf2/NF-κB) signaling. Male albino mice were assigned into five groups as control, vehicle, and LFND (2.5, 5, and 10 mg/kg). We investigated cardiac enzymes, histopathology, and the mRNA expression of Nrf2, NF-κB, BAX, and tumor necrosis factor-α (TNF-α). The bioinformatic study identified the interaction between LFND and Nrf2/NF-κB signaling; this was confirmed by amelioration in mRNA expression (0.5- to 0.34-fold decrease in Nrf2 and 2.6- to 4.61-fold increases in NF-κB genes) and increased (1.76- and 2.625-fold) serum creatine kinase (CK) and 1.38- and 2.33-fold increases in creatine kinase-MB (CK-MB). Histopathological results confirmed the dose-dependent effects of LFND on cardiac muscle structure in the form of cytoplasmic, nuclear, and vascular changes in addition to increased collagen deposits and apoptosis which were increased compared to controls especially with LFND 10 mg/kg. The current study elicits the dose-dependent cardiac injury induced by LFND administration and highlights, for the first time, dysregulation in Nrf2/NF-κB signaling.
Collapse
Affiliation(s)
- Abeer A Rahman
- Department of Histology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Ann Hegazy
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Lamiaa M Elabbasy
- Department of Medical Biochemistry & Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Basic Medical Sciences, College of Medicine, Almaarefa University, Riyadh, Saudi Arabia
| | - Mohamed Z Shoaeir
- Department of Rheumatology and Rehabilitation, Al-Azhar Asyut Faculty of Medicine for Men, Asyut, Egypt
| | - Tarek M Abdel-Aziz
- Department of Rheumatology and Rehabilitation, Al-Azhar Asyut Faculty of Medicine for Men, Asyut, Egypt
| | - Awad S Abbas
- Department of Rheumatology and Rehabilitation, Al-Azhar Asyut Faculty of Medicine for Men, Asyut, Egypt
| | - Heba W Z Khella
- Department of Clinical Education, Canadian Memorial Chiropractic College, Toronto, Canada
| | - Amira H Eltrawy
- Department of Anatomy and Embryology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
- Department of Anatomy, Faculty of Medicine, University of Tabuk, Tabuk, Saudi Arabia
| | - Reem Alshaman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Sheka Yagub Aloyouni
- Research Department, Natural and Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Afaf A Aldahish
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Sawsan A Zaitone
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
18
|
Najafi N, Barangi S, Moosavi Z, Aghaee-Bakhtiari SH, Mehri S, Karimi G. Melatonin Attenuates Arsenic-Induced Neurotoxicity in Rats Through the Regulation of miR-34a/miR-144 in Sirt1/Nrf2 Pathway. Biol Trace Elem Res 2024; 202:3163-3179. [PMID: 37853305 DOI: 10.1007/s12011-023-03897-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023]
Abstract
Arsenic (As) exposure is known to cause several neurological disorders through various molecular mechanisms such as oxidative stress, apoptosis, and autophagy. In the current study, we assessed the effect of melatonin (Mel) on As-induced neurotoxicity. Thirty male Wistar rat were treated daily for 28 consecutive days. As (15 mg/kg, gavage) and Mel (10 and 20 mg/kg, i.p.) were administered to rats. Morris water maze test was done to evaluate learning and memory impairment in training days and probe trial. Oxidative stress markers including MDA and GSH levels, SOD activity, and HO-1 levels were measured. Besides, the levels of apoptosis (caspase 3, Bax/Bcl2 ratio) and autophagy markers (Sirt1, Beclin-1, and LC3 II/I ratio) as well as the expression of miR-144 and miR-34a in cortex tissue were determined. As exposure disturbed learning and memory in animals and Mel alleviated these effects. Also, Mel recovered cortex pathological damages and oxidative stress induced by As. Furthermore, As increased the levels of apoptosis and autophagy proteins in cortex, while Mel (20 mg/kg) decreased apoptosis and autophagy. Also, Mel increased the expression of miR-144 and miR-34a which inhibited by As. In conclusion, Mel administration attenuated As-induced neurotoxicity through anti-oxidative, anti-apoptotic, and anti-autophagy mechanisms, which may be recommended as a therapeutic target for neurological disorders.
Collapse
Affiliation(s)
- Nahid Najafi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samira Barangi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Moosavi
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soghra Mehri
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Abu-Risha SE, Sokar SS, Elzorkany KE, Elsisi AE. Donepezil and quercetin alleviate valproate-induced testicular oxidative stress, inflammation and apoptosis: Imperative roles of AMPK/SIRT1/ PGC-1α and p38-MAPK/NF-κB/ IL-1β signaling cascades. Int Immunopharmacol 2024; 134:112240. [PMID: 38744177 DOI: 10.1016/j.intimp.2024.112240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/23/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
The mounting evidence of valproate-induced testicular damage in clinical settings is alarming, especially for men taking valproate (VPA) for long-term or at high doses. Both donepezil (DON) and quercetin (QUE) have promising antioxidant, anti-inflammatory, and anti-apoptotic effects. Therefore, this study aimed to determine whether DON, QUE, and their combination could mitigate VPA-induced testicular toxicity and unravel the mechanisms underlying their protective effect. In this study, male albino rats were randomly categorized into six equal groups: control, VPA (500 mg/kg, I.P., for 14 days), DON (3 and 5 mg/kg), QUE (50 mg/kg), and DON 3 + QUE combination groups. The DON and QUE treatments were administered orally for 7 consecutive days before VPA administration and then concomitantly with VPA for 14 days. VPA administration disrupted testicular function by altering testicular architecture, ultrastructure, reducing sperm count, viability, and serum testosterone levels. Additionally, VPA triggered oxidative damage, inflammatory, and apoptotic processes and suppressed the AMPK/SIRT1/PGC-1α signaling cascade. Pretreatment with DON, QUE, and their combination significantly alleviated histological and ultrastructure damage caused by VPA and increased the serum testosterone level, sperm count, and viability. They also suppressed the oxidative stress by reducing testicular MDA content and elevating SOD activity. In addition, they reduced the inflammatory response by suppressing IL-1β level, NF-κB, and the p38-MAPK expression as well as inhibiting apoptosis by diminishing caspase-3 and increasing Bcl-2 expression. These novel protective effects were mediated by upregulating AMPK/SIRT1/PGC-1α signaling cascade. In conclusion, these findings suggest that DON, QUE, and their combination possess potent protective effects against VPA-induced testicular toxicity.
Collapse
Affiliation(s)
- Sally E Abu-Risha
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Samia S Sokar
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Kawthar E Elzorkany
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Alaa E Elsisi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
20
|
Abu-Baih RH, Abu-Baih DH, Abdel-Hafez SMN, Fathy M. Activation of SIRT1/Nrf2/HO-1 and Beclin-1/AMPK/mTOR autophagy pathways by eprosartan ameliorates testicular dysfunction induced by testicular torsion in rats. Sci Rep 2024; 14:12566. [PMID: 38822026 PMCID: PMC11143266 DOI: 10.1038/s41598-024-62740-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 05/21/2024] [Indexed: 06/02/2024] Open
Abstract
Testicular torsion carries the ominous prospect of inducing acute scrotal distress and the perilous consequence of testicular atrophy, necessitating immediate surgical intervention to reinstate vital testicular perfusion, notwithstanding the paradoxical detrimental impact of reperfusion. Although no drugs have secured approval for this urgent circumstance, antioxidants emerge as promising candidates. This study aspires to illustrate the influence of eprosartan, an AT1R antagonist, on testicular torsion in rats. Wistar albino rats were meticulously separated into five groups, (n = 6): sham group, eprosartan group, testicular torsion-detorsion (T/D) group, and two groups of T/D treated with two oral doses of eprosartan (30 or 60 mg/kg). Serum testosterone, sperm analysis and histopathological examination were done to evaluate spermatogenesis. Oxidative stress markers were assessed. Bax, BCL-2, SIRT1, Nrf2, HO-1 besides cleaved caspase-3 testicular contents were estimated using ELISA or qRT-PCR. As autophagy markers, SQSTM-1/p62, Beclin-1, mTOR and AMPK were investigated. Our findings highlight that eprosartan effectively improved serum testosterone levels, testicular weight, and sperm count/motility/viability, while mitigating histological irregularities and sperm abnormalities induced by T/D. This recovery in testicular function was underpinned by the activation of the cytoprotective SIRT1/Nrf2/HO-1 axis, which curtailed testicular oxidative stress, indicated by lowering the MDA content and increasing GSH content. In terms of apoptosis, eprosartan effectively countered apoptotic processes by decreasing cleaved caspase-3 content, suppressing Bax and stimulating Bcl-2 gene expression. Simultaneously, it reactivated impaired autophagy by increasing Beclin-1 expression, decreasing the expression of SQSTM-1/p62 and modulate the phosphorylation of AMPK and mTOR proteins. Eprosartan hold promise for managing testicular dysfunction arising from testicular torsion exerting antioxidant, pro-autophagic and anti-apoptotic effect via the activation of SIRT1/Nrf2/HO-1 as well as Beclin-1/AMPK/mTOR pathways.
Collapse
Affiliation(s)
- Rania H Abu-Baih
- Faculty of Pharmacy, Drug Information Center, Minia University, Minia, 61519, Egypt
| | - Dalia H Abu-Baih
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Deraya University, Minia, 61111, Egypt
- Deraya Center for Scientific Research, Deraya University, Minia, 61111, Egypt
| | | | - Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt.
| |
Collapse
|
21
|
Wang Y, Chu Y, Dai H, Zheng Y, Chen R, Zhou C, Zhong Y, Zhan C, Luo J. Protective role of pretreatment with Anisodamine against sepsis-induced diaphragm atrophy via inhibiting JAK2/STAT3 pathway. Int Immunopharmacol 2024; 133:112133. [PMID: 38652962 DOI: 10.1016/j.intimp.2024.112133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
There is an increasing tendency for sepsis patients to suffer from diaphragm atrophy as well as mortality. Therefore, reducing diaphragm atrophy could benefit sepsis patients' prognoses. Studies have shown that Anisodamine (Anis) can exert antioxidant effects when blows occur. However, the role of Anisodamine in diaphragm atrophy in sepsis patients has not been reported. Therefore, this study investigated the antioxidant effect of Anisodamine in sepsis-induced diaphragm atrophy and its mechanism. We used cecal ligation aspiration (CLP) to establish a mouse septic mode and stimulated the C2C12 myotube model with lipopolysaccharide (LPS). After treatment with Anisodamine, we measured the mice's bodyweight, diaphragm weight, fiber cross-sectional area and the diameter of C2C12 myotubes. The malondialdehyde (MDA) levels in the diaphragm were detected using the oxidative stress kit. The expression of MuRF1, Atrogin1 and JAK2/STAT3 signaling pathway components in the diaphragm and C2C12 myotubes was measured by RT-qPCR and Western blot. The mean fluorescence intensity of ROS in C2C12 myotubes was measured by flow cytometry. Meanwhile, we also measured the levels of Drp1 and Cytochrome C (Cyt-C) in vivo and in vitro by Western blot. Our study revealed that Anisodamine alleviated the reduction in diaphragmatic mass and the loss of diaphragmatic fiber cross-sectional area and attenuated the atrophy of the C2C12 myotubes by inhibiting the expression of E3 ubiquitin ligases. In addition, we observed that Anisodamine inhibited the JAK2/STAT3 signaling pathway and protects mitochondrial function. In conclusion, Anisodamine alleviates sepsis-induced diaphragm atrophy, and the mechanism may be related to inhibiting the JAK2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yurou Wang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Chu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongkai Dai
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingfang Zheng
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renyu Chen
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenchen Zhou
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanxia Zhong
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengye Zhan
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinlong Luo
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
22
|
Tian A, Zheng Y, Li H, Zhang Z, Du L, Huang X, Sun L, Wu H. Eicosapentaenoic acid activates the P62/KEAP1/NRF2 pathway for the prevention of diabetes-associated cognitive dysfunction. Food Funct 2024; 15:5251-5271. [PMID: 38680120 DOI: 10.1039/d4fo00774c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Diabetes-associated cognitive dysfunction (DCD) is a severe complication of diabetes mellitus (DM), threatening the life quality of the diabetic population. However, there is still a lack of effective approaches for its intervention. Eicosapentaenoic acid (EPA) is an omega-3 polyunsaturated fatty acid that was not previously investigated for its effect on DCD. In this study, EPA was found to improve DCD in a mouse model of type 2 DM (T2DM) induced by streptozotocin and a high-fat diet, exhibiting profound protective effects on cognitive dysfunction, neuronal loss, and cerebral oxidative stress and inflammation. While EPA did not attenuate advanced glycation end product-induced neuron injury, we hypothesized that EPA might protect neurons by regulating microglia polarization, the effect of which was confirmed by the co-culture of neurons and lipopolysaccharide-stimulated microglia. RNA sequencing identified nuclear factor-erythroid-2-related factor 2 (NRF2) antioxidant signaling as a major target of EPA in microglia. Mechanistically, EPA increased sequestosome-1 (SQSTM1 or P62) levels that might structurally inhibit Kelch-like ECH associated protein 1 (KEAP1), leading to nuclear translocation of NRF2. P62 and NRF2 predominantly mediated EPA's effect since the knockdown of P62 or NRF2 abolished EPA's protective effect on microglial oxidative stress and inflammation and sequential neuron injuries. Moreover, the regulation of P62/KEPA1/NRF2 axes by EPA was confirmed in the hippocampi of diabetic mice. The present work presents EPA as an effective nutritional approach and microglial P62/KEAP1/NRF2 as molecular targets for the intervention of DCD.
Collapse
Affiliation(s)
- Ao Tian
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| | - Yan Zheng
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
| | - Lei Du
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| | - Xiaoli Huang
- Department of Nutrition, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Rd., Jinan, Shandong 250012, China.
| | - Lei Sun
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Rd., Jinan, Shandong 250012, China.
| | - Hao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Rd., Jinan, Shandong 250012, China
- Research Center of Translational Medicine, Jinan Central Hospital, Shandong University, 105 Jiefang Rd., Jinan, Shandong 250013, China.
| |
Collapse
|
23
|
Pang S, Han B, Wu P, Yang X, Liu Y, Li J, Lv Z, Zhang Z. Resveratrol alleviates inorganic arsenic-induced ferroptosis in chicken brain via activation of the Nrf2 signaling pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 201:105885. [PMID: 38685251 DOI: 10.1016/j.pestbp.2024.105885] [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: 02/23/2024] [Revised: 03/09/2024] [Accepted: 03/23/2024] [Indexed: 05/02/2024]
Abstract
Inorganic arsenic (iAs) is a well-recognized environmental pollutant that induces severe brain injury in humans and animals. The antioxidant, anti-inflammatory, and anti-ferroptotic effects of resveratrol (Res) were demonstrated in multiple animal experiments. In order to investigate the protective effect of Res on iAs-induced chicken brain injury, the 40 chickens (19-d-old, female) brain injury model was established by oral administration of iAs (30 mg/L NaAsO2) for 6 weeks. All chickens had free access to both food and water during the experiment. The biochemical indices, hematoxylin-eosin staining, and related protein levels of oxidative stress, inflammation and ferroptosis were then determined. Our results indicated that Res (1000 mg/kg) alleviated the iAs-induced brain injury after 6 weeks of oral administration, primarily by reducing the interleukin-1β mRNA expression and nuclear factor kappa B and malondialdehyde level, and increasing the antioxidant enzyme activity and the mRNA expression of nuclear factor erythroid 2-related factor 2 (Nrf2). Taken together, our study demonstrates that Res effectively inhibits iAs-induced oxidative stress and ferroptosis by mediating the Nrf2 signaling pathway, thereby alleviating iAs-induced brain injury in chickens. This is the first time that the amelioration effects of Res on the iAs-induced brain have been investigated from multiple perspectives.
Collapse
Affiliation(s)
- Shan Pang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Biqi Han
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Xu Yang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Yunfeng Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Jiayi Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Zhanjun Lv
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China.
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China.
| |
Collapse
|
24
|
Khater SI, El-Emam MMA, Abdellatif H, Mostafa M, Khamis T, Soliman RHM, Ahmed HS, Ali SK, Selim HMRM, Alqahtani LS, Habib D, Metwally MMM, Alnakhli AM, Saleh A, Abdelfattah AM, Abdelnour HM, Dowidar MF. Lipid nanoparticles of quercetin (QU-Lip) alleviated pancreatic microenvironment in diabetic male rats: The interplay between oxidative stress - unfolded protein response (UPR) - autophagy, and their regulatory miRNA. Life Sci 2024; 344:122546. [PMID: 38462227 DOI: 10.1016/j.lfs.2024.122546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/20/2024] [Accepted: 03/04/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Autophagy is a well-preserved mechanism essential in minimizing endoplasmic reticulum stress (ER)-related cell death. Defects in β-cell autophagy have been linked to type 1 diabetes, particularly deficits in the secretion of insulin, boosting ER stress sensitivity and possibly promoting pancreatic β-cell death. Quercetin (QU) is a potent antioxidant and anti-diabetic flavonoid with low bioavailability, and the precise mechanism of its anti-diabetic activity is still unknown. Aim This study aimed to design an improved bioavailable form of QU (liposomes) and examine the impact of its treatment on the alleviation of type 1 diabetes induced by STZ in rats. METHODS Seventy SD rats were allocated into seven equal groups 10 rats of each: control, STZ, STZ + 3-MA, STZ + QU-Lip, and STZ + 3-MA + QU-Lip. Fasting blood glucose, insulin, c-peptide, serum IL-6, TNF-α, pancreatic oxidative stress, TRAF-6, autophagy, endoplasmic reticulum stress (ER stress) markers expression and their regulatory microRNA (miRNA) were performed. As well as, docking analysis for the quercetin, ER stress, and autophagy were done. Finally, the histopathological and immunohistochemical analysis were conducted. SIGNIFICANCE QU-Lip significantly decreased glucose levels, oxidative, and inflammatory markers in the pancreas. It also significantly downregulated the expression of ER stress and upregulated autophagic-related markers. Furthermore, QU-Lip significantly ameliorated the expression of several MicroRNAs, which both control autophagy and ER stress signaling pathways. However, the improvement of STZ-diabetic rats was abolished upon combination with an autophagy inhibitor (3-MA). The findings suggest that QU-Lip has therapeutic promise in treating type 1 diabetes by modulating ER stress and autophagy via an epigenetic mechanism.
Collapse
Affiliation(s)
- Safaa I Khater
- Department of Biochemistry and Molecular Biology, Zagazig University, Zagazig 44511, Egypt.
| | | | - Hussein Abdellatif
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Sultanate of Oman; Human Anatomy and Embryology Department, Faculty of Medicine, Mansoura University, Egypt
| | - Mahmoud Mostafa
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt; Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt.
| | | | - Heba S Ahmed
- Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Sahar K Ali
- Department of Clinical Pharmacology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Heba Mohammed Refat M Selim
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, AlMaarefa University, Diriyah 13713, Riyadh, Saudi Arabia; Microbiology and Immunology Department, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 35527, Egypt
| | - Leena S Alqahtani
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah 23445, Saudi Arabia
| | - Doaa Habib
- Department of Biochemistry and Molecular Biology, Zagazig University, Zagazig 44511, Egypt
| | - Mohamed M M Metwally
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt; Department of pathology and clinical pathology, faculty of veterinary medicine, King Salman international University, Ras sidr, Egypt
| | - Anwar M Alnakhli
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, 84428, Riyadh 11671, Saudi Arabia
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, 84428, Riyadh 11671, Saudi Arabia
| | | | - Hanim M Abdelnour
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed F Dowidar
- Department of Biochemistry and Molecular Biology, Zagazig University, Zagazig 44511, Egypt
| |
Collapse
|
25
|
Yang X, Guo C, Yu L, Lv Z, Li S, Zhang Z. Dendrobium officinale polysaccharide alleviates thiacloprid-induced kidney injury in quails via activating the Nrf2/HO-1 pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:2655-2666. [PMID: 38224485 DOI: 10.1002/tox.24137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/14/2023] [Accepted: 12/29/2023] [Indexed: 01/17/2024]
Abstract
Thiacloprid (THI) is a neonicotinoid insecticide, and its wide-ranging use has contributed to severe environmental and health problems. Dendrobium officinale polysaccharide (DOP) possesses multiple biological activities such as antioxidant and antiapoptosis effect. Although present research has shown that THI causes kidney injury, the exact molecular mechanism and treatment of THI-induced kidney injury remain unclear. The study aimed to investigate if DOP could alleviate THI-induced kidney injury and identify the potential molecular mechanism in quails. In this study, Japanese quails received DOP (200 mg/kg) daily with or without THI (4 mg/kg) exposure for 42 days. Our results showed that DOP improved hematological changes, biochemical indexes, and nephric histopathological changes induced by THI. Meanwhile, THI exposure caused oxidative stress, apoptosis, and autophagy. Furthermore, THI and DOP cotreatment significantly activated the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) pathway, restored antioxidant enzyme activity, and reduced apoptosis and autophagy in quail kidneys. In summary, our study demonstrated that DOP mitigated THI-mediated kidney injury was associated with oxidative stress, apoptosis, and autophagy via activation of the Nrf2/HO-1 signaling pathway in quails.
Collapse
Affiliation(s)
- Xu Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Changming Guo
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lu Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhanjun Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
26
|
Abubakar MG, Agbon AN, Musa SA, Hamman WO, Oladele SB. Biochemical, morphological and molecular assessments of n butanol fraction of Phoenix dactylifera L. following exposure to inorganic mercury on the liver of Wistar rats. Lab Anim Res 2024; 40:15. [PMID: 38641806 PMCID: PMC11027370 DOI: 10.1186/s42826-024-00203-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/06/2024] [Accepted: 04/13/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Mercury chloride (HgCl2) damages tissues it comes in contact with in sufficient concentration. This study evaluated the protective effects of n-butanol fraction of Phoenix dactylifera (BFPD) on mercury-triggered liver toxicity in Wistar rats. 25 male rats were divided into 5 groups of 5 rats each. Group I was administered 2 ml/kg of distilled water; group II was administered 5 mg/kg of HgCl2; group III was administered 500 mg/kg of BFPD + 5 mg/kg of HgCl2; group IV was administered 1000 mg/kg of BFPD + 5 mg/kg of HgCl2, while group V was administered 100 mg/kg of silymarin + 5 mg/kg of HgCl2. orally for 2 weeks. The rats were euthanized and liver tissue blood samples were collected for histological, histochemical, stereological, immunohistochemical, molecular, and biochemical studies. RESULTS The results revealed that HgCl2 induced oxidative stress in the rats evident by histoarchitectural distortions and altered levels of liver enzymes, proteins, and oxidative stress biomarkers when compared to the control. However, BFPD treatment restored these changes. Glutathione peroxidase levels decreased (p < 0.05) in the HgCl2-treated group when compared to the control and BFPD-treated groups. HgCl2 group revealed reduced reactivity with histochemical and immunohistochemical stains (Masson's Trichrome and B cell Lymphoma 2) when compared to the control, with a significant decrease in quantified liver Bcl-2 stain intensity when compared to the silymarin-treated group. BFPD administration revealed normal staining intensity comparable to the control. HgCl2 administration revealed a remarked decrease in the number of hepatocytes when compared to the control, BFPD, and silymarin groups. BFPD preserved (p < 0.05) the stereological features when compared to the HgCl2-treated group. GPx activity in the liver decreased (p < 0.05) with HgCl2 administration when compared to the control and silymarin-treated groups. BFPD attenuated GPx gene activity to levels similar to the control indicating some level of amelioration against HgCl2-induced toxicity. CONCLUSIONS The ability of BFPD to mitigate HgCl2 triggered liver alterations could be attributed to the antioxidant property of its flavonoid content. Therefore, BFPD may be a potential candidate for treating and managing liver-induced mercury intoxication.
Collapse
Affiliation(s)
- Musa Garba Abubakar
- Microscopy and Stereology Research Unit, Department of Human Anatomy, Ahmadu Bello University, Zaria, Nigeria.
- Nigerian Defence Academy, Kaduna, Nigeria.
| | - A N Agbon
- Microscopy and Stereology Research Unit, Department of Human Anatomy, Ahmadu Bello University, Zaria, Nigeria
| | - S A Musa
- Microscopy and Stereology Research Unit, Department of Human Anatomy, Ahmadu Bello University, Zaria, Nigeria
| | - W O Hamman
- Microscopy and Stereology Research Unit, Department of Human Anatomy, Ahmadu Bello University, Zaria, Nigeria
| | - S B Oladele
- Department of Veterinary Pathology, Faculty of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| |
Collapse
|
27
|
Sadek MA, Rabie MA, El Sayed NS, Sayed HM, Kandil EA. Neuroprotective effect of curcumin against experimental autoimmune encephalomyelitis-induced cognitive and physical impairments in mice: an insight into the role of the AMPK/SIRT1 pathway. Inflammopharmacology 2024; 32:1499-1518. [PMID: 38112964 PMCID: PMC11006778 DOI: 10.1007/s10787-023-01399-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/18/2023] [Indexed: 12/21/2023]
Abstract
Multiple sclerosis (MS) is an incurable chronic neurodegenerative disease where autoimmunity, oxidative stress, and neuroinflammation collaboration predispose myelin sheath destruction. Interestingly, curcumin, a natural polyphenol, showed a neuroprotective effect in numerous neurodegenerative diseases, including MS. Nevertheless, the influence of curcumin against MS-induced cognitive impairment is still vague. Hence, we induced experimental autoimmune encephalomyelitis (EAE) in mice using spinal cord homogenate (SCH) and complete Freund's adjuvant, which eventually mimic MS. This study aimed not only to evaluate curcumin efficacy against EAE-induced cognitive and motor dysfunction, but also to explore a novel mechanism of action, by which curcumin exerts its beneficial effects in MS. Curcumin (200 mg/kg/day) efficacy was evaluated by behavioral tests, histopathological examination, and biochemical tests. Concisely, curcumin amended EAE-induced cognitive and motor impairments, as demonstrated by the behavioral tests and histopathological examination of the hippocampus. Interestingly, curcumin activated the adenosine monophosphate (AMP)-activated protein kinase/silent mating type information regulation 2 homolog 1 (AMPK/SIRT1) axis, which triggered cyclic AMP response element-binding protein/brain-derived neurotrophic factor/myelin basic protein (CREB/BDNF/MBP) pathway, hindering demyelination of the corpus callosum. Furthermore, AMPK/SIRT1 activation augmented nuclear factor erythroid 2-related factor 2 (Nrf2), a powerful antioxidant, amending EAE-induced oxidative stress. Additionally, curcumin abolished EAE-induced neuroinflammation by inhibiting Janus kinase 2 /signal transducers and activators of transcription 3 (JAK2/STAT3) axis, by various pathways, including AMPK/SIRT1 activation. JAK2/STAT3 inhibition halts inflammatory cytokines synthesis. In conclusion, curcumin's neuroprotective effect in EAE is controlled, at least in part, by AMPK/SIRT1 activation, which ultimately minimizes EAE-induced neuronal demyelination, oxidative stress, and neuroinflammation.
Collapse
Affiliation(s)
- Mohamed A Sadek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Helmy M Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Esraa A Kandil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
28
|
Lin HW, Lee HL, Shen TJ, Ho MT, Lee YJ, Wang I, Lin CP, Chang YY. Pb(NO 3 ) 2 induces cell apoptosis through triggering of reactive oxygen species accumulation and disruption of mitochondrial function via SIRT3/SOD2 pathways. ENVIRONMENTAL TOXICOLOGY 2024; 39:1294-1302. [PMID: 37948429 DOI: 10.1002/tox.24019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/26/2023] [Accepted: 10/07/2023] [Indexed: 11/12/2023]
Abstract
Lead (Pb) is nonbiodegradable and toxic to the lungs. To investigate the potential mechanisms of Pb-induced reactive oxygen species (ROS) accumulation and cell death in the lungs, human non-small lung carcinoma H460 cells were stimulated with Pb(NO3 )2 in this study. The results showed that Pb(NO3 )2 stimulation increased cell death by inducing cell apoptosis which showed a reduced Bcl-2 expression and an enhanced caspase 3 activation. Pb(NO3 )2 also caused the production of H2 O2 in H460 cells that triggering the buildup of ROS and mitochondrial membrane potential loss. We found that Pb(NO3 )2 modulates oxidoreductive activity through reduced the glutathione-disulfide reductase and glutathione levels in Pb(NO3 )2 -exposed H460 cells. Furthermore, the superoxide dismutase (SOD) upstream molecule sirtuin 3 (SIRT3) was increased with Pb(NO3 )2 dose. Collectively, these results demonstrate that Pb(NO3 )2 promotes lung cell death through SIRT3/SOD-mediated ROS accumulation and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Hui-Wen Lin
- Department of Optometry, Asia University, Taichung, Taiwan
| | - Hsiang-Lin Lee
- Department of Surgery, School of Medicine, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung, Taiwan
| | - Ting-Jing Shen
- Department of Microbiology and Immunology, School of Medicine, Chung Shan Medical University, and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Meng-Ting Ho
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Ju Lee
- Department of Pathology, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Inga Wang
- Rehabilitation Sciences & Technology, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin, USA
| | - Ching-Pin Lin
- Division of Hematology and Gastroenterology, Department of internal Medicine, School of Medicine, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung, Taiwan
| | - Yuan-Yen Chang
- Department of Surgery, School of Medicine, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung, Taiwan
- Department of Microbiology and Immunology, School of Medicine, Chung Shan Medical University, and Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
29
|
Bayav I, Darendelioğlu E, Caglayan C. 18β-Glycyrrhetinic acid exerts cardioprotective effects against BPA-induced cardiotoxicity through antiapoptotic and antioxidant mechanisms. J Biochem Mol Toxicol 2024; 38:e23655. [PMID: 38348715 DOI: 10.1002/jbt.23655] [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: 05/30/2023] [Revised: 12/02/2023] [Accepted: 01/11/2024] [Indexed: 02/15/2024]
Abstract
Bisphenol A (BPA) is a synthetic environmental pollutant widely used in industry, as well as is an endocrine disrupting chemicals and has a toxic effects on heart tissue. The aim of this study is to reveal the cardioprotective effects of 18β-glycyrretinic acid (GA) against BPA-induced cardiotoxicity in rats. In this study, 40 male rats were used and five different groups (each group includes eight rats) were formed. The rats were applied BPA (250 mg/kg b.w.) alone or with GA (50 and 100 mg/kg b.w.) for 14 days. Rats were killed on Day 15 and heart tissues were taken for analysis. GA treatment decreased serum lactate dehydrogenase and creatine kinase MB levels, reducing BPA-induced heart damage. GA treatment showed ameliorative effects against lipid peroxidation and oxidative stress caused by BPA by increasing the antioxidant enzyme activities (glutathione peroxidase, superoxide dismutase, and catalase) and GSH level of the heart tissue and decreasing the MDA level. In addition, GA showed antiapoptotic effect by increasing Bcl-2, procaspase-3, and -9 protein expression levels and decreasing Bax, cytochrome c, and P53 protein levels in heart tissue. As a result, it was found that GA has cardioprotective effects on heart tissue by exhibiting antioxidant and antiapoptotic effects against heart damage caused by BPA, an environmental pollutant. Thus, it was supported that GA could be a potential cardioprotective agent.
Collapse
Affiliation(s)
- Ibrahim Bayav
- Department of Medical Biology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Ekrem Darendelioğlu
- Department of Molecular Biology and Genetics, Faculty of Science and Literature, Bingol University, Bingol, Turkey
| | - Cuneyt Caglayan
- Department of Medical Biochemistry, Faculty of Medicine, Bilecik Seyh Edebali University, Bilecik, Turkey
| |
Collapse
|
30
|
Kim M, Jee SC, Sung JS. Hepatoprotective Effects of Flavonoids against Benzo[a]Pyrene-Induced Oxidative Liver Damage along Its Metabolic Pathways. Antioxidants (Basel) 2024; 13:180. [PMID: 38397778 PMCID: PMC10886006 DOI: 10.3390/antiox13020180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Benzo[a]pyrene (B[a]P), a highly carcinogenic polycyclic aromatic hydrocarbon primarily formed during incomplete organic matter combustion, undergoes a series of hepatic metabolic reactions once absorbed into the body. B[a]P contributes to liver damage, ranging from molecular DNA damage to the onset and progression of various diseases, including cancer. Specifically, B[a]P induces oxidative stress via reactive oxygen species generation within cells. Consequently, more research has focused on exploring the underlying mechanisms of B[a]P-induced oxidative stress and potential strategies to counter its hepatic toxicity. Flavonoids, natural compounds abundant in plants and renowned for their antioxidant properties, possess the ability to neutralize the adverse effects of free radicals effectively. Although extensive research has investigated the antioxidant effects of flavonoids, limited research has delved into their potential in regulating B[a]P metabolism to alleviate oxidative stress. This review aims to consolidate current knowledge on B[a]P-induced liver oxidative stress and examines the role of flavonoids in mitigating its toxicity.
Collapse
Affiliation(s)
| | | | - Jung-Suk Sung
- Department of Life Science, Dongguk University-Seoul, Goyang 10326, Republic of Korea; (M.K.); (S.-C.J.)
| |
Collapse
|
31
|
Sharma S, Mehan S, Khan Z, Gupta GD, Narula AS. Icariin prevents methylmercury-induced experimental neurotoxicity: Evidence from cerebrospinal fluid, blood plasma, brain samples, and in-silico investigations. Heliyon 2024; 10:e24050. [PMID: 38226245 PMCID: PMC10788811 DOI: 10.1016/j.heliyon.2024.e24050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/29/2023] [Accepted: 01/02/2024] [Indexed: 01/17/2024] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a fatal neurodegenerative disease that causes significant neurodegeneration. Methylmercury (MeHg+) is a neurotoxin that induces axonal neurodegeneration and motor nerve degeneration by destroying oligodendrocytes, degenerating white matter, inducing apoptosis, excitotoxicity, and reducing myelin basic protein (MBP). This study examines the inhibition of SIRT-1 (silence information regulator 1), Nrf-2 (nuclear factor E2-related factor 2), HO-1 (heme oxygenase 1), and TDP-43 (TAR-DNA-binding protein 43) accumulation in the context of ALS, as well as the modulation of these proteins by icariin (15 and 30 mg/kg, orally), a glycoside flavonoid with neuroprotective properties. Neuroprotective icariin activates SIRT-1, Nrf-2, and HO-1, mitigating inflammation and neuronal injury in neurodegenerative disorders. In-vivo and in-silico testing of experimental ALS models confirmed icariin efficacy in modulating these cellular targets. The addition of sirtinol 10 mg/kg, an inhibitor of SIRT-1, helps determine the effectiveness of icariin. In this study, we also examined neurobehavioral, neurochemical, histopathological, and LFB (Luxol fast blue) markers in various biological samples, including Cerebrospinal fluid (CSF), blood plasma, and brain homogenates (Cerebral Cortex, Hippocampus, Striatum, mid-brain, and Cerebellum). These results demonstrate that the administration of icariin ameliorates experimental ALS and that the mechanism underlying these benefits is likely related to regulating the SIRT-1, Nrf-2, and HO-1 signaling pathways.
Collapse
Affiliation(s)
- Sarthak Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India), Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India), Moga, Punjab, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India), Moga, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy (Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab, 144603, India), Moga, Punjab, India
| | | |
Collapse
|
32
|
Han X, Wang H, Du F, Zeng X, Guo C. Nrf2 for a key member of redox regulation: A novel insight against myocardial ischemia and reperfusion injuries. Biomed Pharmacother 2023; 168:115855. [PMID: 37939614 DOI: 10.1016/j.biopha.2023.115855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/21/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023] Open
Abstract
Nuclear factor erythroid-2 related factor 2 (Nrf2), a nuclear transcription factor, modulates genes responsible for antioxidant responses against toxic and oxidative stress to maintain redox homeostasis and participates in varieties of cellular processes such as metabolism and inflammation during myocardial ischemia and reperfusion injuries (MIRI). The accumulation of reactive oxygen species (ROS) from damaged mitochondria, xanthine oxidase, NADPH oxidases, and inflammation contributes to depraved myocardial ischemia and reperfusion injuries. Considering that Nrf2 played crucial roles in antagonizing oxidative stress, it is reasonable to delve into the up or down-regulated molecular mechanisms of Nrf2 in the progression of MIRI to provide the possibility of new therapeutic medicine targeting Nrf2 in cardiovascular diseases. This review systematically describes the generation of ROS, the regulatory metabolisms of Nrf2 as well as several natural or synthetic compounds activating Nrf2 during MIRI, which might provide novel insights for the anti-oxidative stress and original ideas targeting Nrf2 for the prevention and treatment in cardiovascular diseases.
Collapse
Affiliation(s)
- Xuejie Han
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China
| | - Hongxia Wang
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China
| | - Fenghe Du
- Department of Geriatrics, Beijing Tiantan Hospital, Capital Medical University, No. 119 South 4th Ring West Road, Fengtai District, Beijing 100070, PR China
| | - Xiangjun Zeng
- Department of Physiology and Pathophysiology, Capital Medical University, No. 10 You An Men Wai Xi Tou Tiao, Fengtai District, Beijing 100069, PR China.
| | - Caixia Guo
- Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 1 Dongjiaomin Lane, Dongcheng District, Beijing 100730, PR China.
| |
Collapse
|
33
|
Ramadan SS, El Zaiat FA, Habashy EA, Montaser MM, Hassan HE, Tharwat SS, El-khadragy M, Abdel Moneim AE, Elshopakey GE, Akabawy AMA. Coenzyme Q10-Loaded Albumin Nanoparticles Protect against Redox Imbalance and Inflammatory, Apoptotic, and Histopathological Alterations in Mercuric Chloride-Induced Hepatorenal Toxicity in Rats. Biomedicines 2023; 11:3054. [PMID: 38002054 PMCID: PMC10669886 DOI: 10.3390/biomedicines11113054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/27/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Exposure to mercuric chloride (HgCl2), either accidental or occupational, induces substantial liver and kidney damage. Coenzyme Q10 (CoQ10) is a natural antioxidant that also has anti-inflammatory and anti-apoptotic activities. Herein, our study aimed to investigate the possible protective effects of CoQ10 alone or loaded with albumin nanoparticles (CoQ10NPs) against HgCl2-induced hepatorenal toxicity in rats. Experimental animals received CoQ10 (10 mg/kg/oral) or CoQ10NPs (10 mg/kg/oral) and were injected intraperitoneally with HgCl2 (5 mg/kg; three times/week) for two weeks. The results indicated that CoQ10NP pretreatment caused a significant decrease in serum liver and kidney function markers. Moreover, lowered MDA and NO levels were associated with an increase in antioxidant enzyme activities (SOD, GPx, GR, and CAT), along with higher GSH contents, in both the liver and kidneys of intoxicated rats treated with CoQ10NPs. Moreover, HgCl2-intoxicated rats that received CoQ10NPs revealed a significant reduction in the hepatorenal levels of TNF-α, IL-1β, NF-κB, and TGF-β, as well as an increase in the hepatic level of the fibrotic marker (α-SMA). Notably, CoQ10NPs counteracted hepatorenal apoptosis by diminishing the levels of Bax and caspase-3 and boosting the level of Bcl-2. The hepatic and renal histopathological findings supported the abovementioned changes. In conclusion, these data suggest that CoQ10, alone or loaded with albumin nanoparticles, has great power in reversing the hepatic and renal tissue impairment induced by HgCl2 via the modulation of hepatorenal oxidative damage, inflammation, and apoptosis. Therefore, this study provides a valuable therapeutic agent (CoQ10NPs) for preventing and treating several HgCl2-induced hepatorenal disorders.
Collapse
Affiliation(s)
- Shimaa S. Ramadan
- Biochemistry Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Farah A. El Zaiat
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Engy A. Habashy
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Mostafa M. Montaser
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Habeba E. Hassan
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Shahinaz S. Tharwat
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Manal El-khadragy
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Ahmed E. Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Gehad E. Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed M. A. Akabawy
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| |
Collapse
|
34
|
Hussain MS, Gupta G, Goyal A, Thapa R, Almalki WH, Kazmi I, Alzarea SI, Fuloria S, Meenakshi DU, Jakhmola V, Pandey M, Singh SK, Dua K. From nature to therapy: Luteolin's potential as an immune system modulator in inflammatory disorders. J Biochem Mol Toxicol 2023; 37:e23482. [PMID: 37530602 DOI: 10.1002/jbt.23482] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/05/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023]
Abstract
Inflammation is an essential immune response that helps fight infections and heal tissues. However, chronic inflammation has been linked to several diseases, including cancer, autoimmune disorders, cardiovascular diseases, and neurological disorders. This has increased interest in finding natural substances that can modulate the immune system inflammatory signaling pathways to prevent or treat these diseases. Luteolin is a flavonoid found in many fruits, vegetables, and herbs. It has been shown to have anti-inflammatory effects by altering signaling pathways in immune cells. This review article discusses the current research on luteolin's role as a natural immune system modulator of inflammatory signaling mechanisms, such as its effects on nuclear factor-kappa B, mitogen-activated protein kinases, Janus kinase/signal transducer and activator of transcription, and inflammasome signaling processes. The safety profile of luteolin and its potential therapeutic uses in conditions linked to inflammation are also discussed. Overall, the data point to Luteolin's intriguing potential as a natural regulator of immune system inflammatory signaling processes. More research is needed to fully understand its mechanisms of action and possible therapeutic applications.
Collapse
Affiliation(s)
- Md Sadique Hussain
- School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, India
| | - Gaurav Gupta
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
- Center for Global Health research (CGHR), Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
| | | | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | | | | | - Vikas Jakhmola
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Manisha Pandey
- Department of Pharmaceutical Sciences, Central University of Haryana, Mahendergarh, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
35
|
Zhu X, Wu Y, Zhang X, Gu W, Ning Z. Stachydrine ameliorates hypoxia reoxygenation injury of cardiomyocyte via enhancing SIRT1-Nrf2 pathway. J Cardiothorac Surg 2023; 18:265. [PMID: 37752609 PMCID: PMC10521545 DOI: 10.1186/s13019-023-02363-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
BACKGROUND Hypoxia/reoxygenation (H/R)-induced cardiomyocyte cell apoptosis is critical in developing myocardial infarction. Stachydrine (STA), an active constituent of Leonurus heterophyllus sweet, could have a protective effect on myocardial H/R injury, which remains unexplored. Therefore, the study aimed to investigate the protective effects and mechanisms of STA on H/R injury of cardiomyocytes. METHODS Rat cardiomyocyte H9c2 cells underwent H/R (hypoxia for 4 h and reoxygenation for 12 h). Cells were pretreated with STA (50 µM) 2 h before H/R. Cardiomyocyte injury was evaluated by CCK-8 assay and lactate dehydrogenase (LDH) release. Apoptosis was assessed by TUNEL staining and caspase-3 activity. Oxidative stress was assessed by lipid oxidation product MDA and a ROS-scavenging enzyme SOD in culture media. Western blot was performed to measure the protein expressions of SIRT1, Nrf2, and heme oxygenase-1 (HO-1). RESULTS STA reversed the decrease in cell viability and increased LDH release in H9c2 cells with the H/R insult. STA significantly suppressed oxidative stress, reduced MDA content, and increased SOD activity in H9c2 cells exposed to H/R. STA reduced apoptosis in H9c2 cells exposed to H/R, as evidenced by the reduced TUNEL positive cells and caspase-3 activity. In addition, STA enhanced SIRT1, Nrf2, and HO-1 protein expression in H/R-stimulated H9c2 cells. SIRT1 and Nrf2 involved the protective effect of STA in H/R-exposed H9c2 cells, as the changes in cell viability and caspase-3 activity by STA can be reversed by SIRT1 inhibitor EX-527 or Nrf2 siRNA. CONCLUSIONS Our data speculated that STA protects H/R injury and inhibits oxidative stress and apoptosis in cardiomyocytes by activation of the SIRT1-Nrf2 pathway.
Collapse
Affiliation(s)
- Xi Zhu
- Department of Cardiology, Shanghai University of Medicine & Health Sciences affiliated Zhoupu Hospital, No.1500 Zhouyuan Road, Pudong New District, Shanghai, 201318, China
| | - Yingbiao Wu
- Department of Cardiology, Shanghai University of Medicine & Health Sciences affiliated Zhoupu Hospital, No.1500 Zhouyuan Road, Pudong New District, Shanghai, 201318, China
| | - Xiaogang Zhang
- Department of Cardiology, Shanghai University of Medicine & Health Sciences affiliated Zhoupu Hospital, No.1500 Zhouyuan Road, Pudong New District, Shanghai, 201318, China
| | - Wei Gu
- Department of Cardiology, Shanghai University of Medicine & Health Sciences affiliated Zhoupu Hospital, No.1500 Zhouyuan Road, Pudong New District, Shanghai, 201318, China
| | - Zhongping Ning
- Department of Cardiology, Shanghai University of Medicine & Health Sciences affiliated Zhoupu Hospital, No.1500 Zhouyuan Road, Pudong New District, Shanghai, 201318, China.
| |
Collapse
|
36
|
Yin H, Ran Z, Luo T, Jin Z, Ma J. BCL-3 Promotes Intracerebral Hemorrhage Progression by Increasing Blood-Brain Barrier Permeability, Inflammation, and Cell Apoptosis via Endoplasmic Reticulum Stress. Mediators Inflamm 2023; 2023:1420367. [PMID: 37736616 PMCID: PMC10511295 DOI: 10.1155/2023/1420367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 09/23/2023] Open
Abstract
Background Intracerebral hemorrhage (ICH) is among the common types of stroke with high mortality and morbidity. Molecular biomarker selection is crucial for ICH diagnosis and treatment. However, the identification of ICH-related biomarkers remains inadequate. Materials and Methods In vivo and in vitro ICH models were generated and transfected with silenced B-cell lymphoma-3 (BCL-3 and siRNA BCL-3), overexpressed BCL-3, and endoplasmic reticulum stress (ERS) agonist (2-CLHA). Hematoxylin-eosin staining and transmission electron microscopy were used to observe the transfected cells. RNA sequencing was performed in vivo on the sham and ICH groups. The blood-brain barrier (BBB) permeability was evaluated by determining Evans blue dye extravasation, transendothelial electrical resistance, and paracellular permeability. Moreover, tight junction-, cell apoptosis-, and endoplasmic reticulum stress- (ERS-) related proteins were evaluated through real-time quantitative PCR, western blotting, immunohistochemistry, and TUNEL staining. The levels of inflammatory cytokines were measured through the enzyme-linked immunosorbent assay. Results RNA-seq revealed that BCL-3 acts as a key player. BCL-3 promotes ICH progression by increasing BBB permeability, ERS, inflammation, and cell apoptosis. Silencing of BCL-3 slows ICH progression by reducing BBB permeability and inflammation and terminating cell apoptosis and ERS in vitro and in vivo. Conclusion Our study identified ICH biomarkers and elucidated the role of BCL-3 in ICH for the first time.
Collapse
Affiliation(s)
- Hao Yin
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| | - Zhongying Ran
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| | - Tao Luo
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| | - Zexin Jin
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| | - Jun Ma
- Department of Neurosurgery, Guizhou Provincial People's Hospital, China
| |
Collapse
|
37
|
Huang W, Zhong Y, Gao B, Zheng B, Liu Y. Nrf2-mediated therapeutic effects of dietary flavones in different diseases. Front Pharmacol 2023; 14:1240433. [PMID: 37767395 PMCID: PMC10520786 DOI: 10.3389/fphar.2023.1240433] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Oxidative stress (OS) is a pathological status that occurs when the body's balance between oxidants and antioxidant defense systems is broken, which can promote the development of many diseases. Nrf2, a redox-sensitive transcription encoded by NFE2L2, is the master regulator of phase II antioxidant enzymes and cytoprotective genes. In this context, Nrf2/ARE signaling can be a compelling target against OS-induced diseases. Recently, natural Nrf2/ARE regulators like dietary flavones have shown therapeutic potential in various acute and chronic diseases such as diabetes, neurodegenerative diseases, ischemia-reperfusion injury, and cancer. In this review, we aim to summarize nrf2-mediated protective effects of flavones in different conditions. Firstly, we retrospected the mechanisms of how flavones regulate the Nrf2/ARE pathway and introduced the mediator role Nrf2 plays in inflammation and apoptosis. Then we review the evidence that flavones modulated Nrf2/ARE pathway to prevent diseases in experimental models. Based on these literature, we found that flavones could regulate Nrf2 expression by mechanisms below: 1) dissociating the binding between Nrf2 and Keap1 via PKC-mediated Nrf2 phosphorylation and P62-mediated Keap1 autophagic degradation; 2) regulating Nrf2 nuclear translocation by various kinases like AMPK, MAPKs, Fyn; 3) decreasing Nrf2 ubiquitination and degradation via activating sirt1 and PI3K/AKT-mediated GSK3 inhibition; and 4) epigenetic alternation of Nrf2 such as demethylation at the promoter region and histone acetylation. In conclusion, flavones targeting Nrf2 can be promising therapeutic agents for various OS-related disorders. However, there is a lack of investigations on human subjects, and new drug delivery systems to improve flavones' treatment efficiency still need to be developed.
Collapse
Affiliation(s)
- Wenkai Huang
- Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yuan Zhong
- Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Botao Gao
- Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Bowen Zheng
- Liaoning Provincial Key Laboratory of Oral Disease, Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Yi Liu
- Liaoning Provincial Key Laboratory of Oral Disease, Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
38
|
Shehnaz SI, Roy A, Vijayaraghavan R, Sivanesan S. Luteolin Mitigates Diabetic Dyslipidemia in Rats by Modulating ACAT-2, PPARα, SREBP-2 Proteins, and Oxidative Stress. Appl Biochem Biotechnol 2023; 195:4893-4914. [PMID: 37103741 DOI: 10.1007/s12010-023-04544-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2023] [Indexed: 04/28/2023]
Abstract
Diabetic dyslipidemia is a crucial link between type-2 diabetes mellitus (T2DM) and atherosclerotic cardiovascular diseases (ASCVD). Natural biologically active substances have been advocated as complementary remedies for ASCVD and T2DM. Luteolin, a flavonoid, exhibits antioxidant, hypolipidemic, and antiatherogenic effects. Hence, we aimed to determine influence of luteolin on lipid homeostasis and hepatic damage in rats with T2DM induced by high-fat-diet (HFD) and streptozotocin (STZ). After being fed HFD for 10 days, male Wistar rats received 40 mg/kg STZ intraperitoneal injection on 11th day. Seventy-two hours later, hyperglycemic rats (fasting glucose > 200 mg/dL) were randomized into groups, and oral hydroxy-propyl-cellulose, atorvastatin (5 mg/kg), or luteolin (50 mg/kg or 100 mg/kg) administered daily, while continuing HFD for 28 days. Luteolin significantly ameliorated dyslipidemia levels and concomitantly improved atherogenic index of plasma in a dose-dependent manner. Increased levels of malondialdehyde and diminished levels of superoxide dismutase, catalase, and glutathione in HFD-STZ-diabetic rats were significantly regulated by luteolin. Luteolin significantly intensified PPARα expression while decreasing expression of acyl-coenzyme A:cholesterol acyltransferase-2 (ACAT-2) and sterol regulatory element binding protein-2 (SREBP-2) proteins. Moreover, luteolin effectively alleviated hepatic impairment in HFD-STZ-diabetic rats to near-normal control levels. The findings of the present study expound mechanisms by which luteolin mitigated diabetic dyslipidemia and alleviated hepatic impairment in HFD-STZ-diabetic rats by amelioration of oxidative stress, modulation of PPARα expression, and downregulation of ACAT-2 and SREBP-2. In conclusion, our results imply that luteolin may be efficacious in management of dyslipidemia in T2DM, and future research may be essential to substantiate our findings.
Collapse
Affiliation(s)
- Syed Ilyas Shehnaz
- Department of Pharmacology, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, Tamil Nadu, India.
| | - Anitha Roy
- Centre for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, Tamil Nadu, India
| | - Rajagopalan Vijayaraghavan
- Department of Research and Development, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, Tamil Nadu, India
| | - Senthilkumar Sivanesan
- Department of Research and Development, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, Tamil Nadu, India
- Department of Biosciences, Institute of Biotechnology, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, Tamil Nadu, India
| |
Collapse
|
39
|
Xing X, Peng J, Zhao J, Shi R, Wang C, Zhang Z, Wang Z, Li Z, Wu Z. Luteolin regulates the distribution and function of organelles by controlling SIRT1 activity during postovulatory oocyte aging. Front Nutr 2023; 10:1192758. [PMID: 37583461 PMCID: PMC10424794 DOI: 10.3389/fnut.2023.1192758] [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: 03/23/2023] [Accepted: 07/04/2023] [Indexed: 08/17/2023] Open
Abstract
The quality of oocytes determines their development competence, which will be rapidly lost if the oocytes are not fertilized at the proper time after ovulation. SIRT1, one of the sirtuin family members, has been proven to protect the quality of oocytes during postovulatory oocyte aging. However, evidence of the effect of SIRT1 on the activity of organelles including the mitochondria, the endoplasmic reticulum (ER), the Golgi apparatus, and the lysosomes in postovulatory aging oocyte is lacking. In this study, we investigated the distribution and function of organelles in postovulatory aged oocytes and discovered abnormalities. Luteolin, which is a natural flavonoid contained in vegetables and fruits, is an activator of SIRT1. When the oocytes were treated with luteolin, the abnormal distribution of mitochondria, ER, and Golgi complex were restored during postovulatory oocyte aging. The ER stress protein GRP78 and the lysosome protein LAMP1 increased, while the mitochondrial membrane potential and the Golgi complex protein GOLPH3 decreased in aged oocytes, and these were restored by luteolin treatment. EX-527, an inhibitor of SIRT1, disrupted the luteolin-mediated normal distribution and function of mitochondria, ER, Golgi apparatus, and lysosomes. In conclusion, we demonstrate that luteolin regulates the distribution and function of mitochondria, ER, Golgi apparatus, and lysosomes during postovulatory oocyte aging by activating SIRT1.
Collapse
Affiliation(s)
- Xupeng Xing
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jingfeng Peng
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jingyu Zhao
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Ruoxi Shi
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Caiqin Wang
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Zihan Zhang
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Zihan Wang
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, China
- College of Second Clinical Medical, Jining Medical University, Jining, China
| | - Zicong Li
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhenfang Wu
- National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, China
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
40
|
Abd El-Emam MM, Mostafa M, Farag AA, Youssef HS, El-Demerdash AS, Bayoumi H, Gebba MA, El-Halawani SM, Saleh AM, Badr AM, El Sayed S. The Potential Effects of Quercetin-Loaded Nanoliposomes on Amoxicillin/Clavulanate-Induced Hepatic Damage: Targeting the SIRT1/Nrf2/NF-κB Signaling Pathway and Microbiota Modulation. Antioxidants (Basel) 2023; 12:1487. [PMID: 37627483 PMCID: PMC10451903 DOI: 10.3390/antiox12081487] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/16/2023] [Accepted: 07/19/2023] [Indexed: 08/27/2023] Open
Abstract
Amoxicillin/clavulanate (Co-Amox), a commonly used antibiotic for the treatment of bacterial infections, has been associated with drug-induced liver damage. Quercetin (QR), a naturally occurring flavonoid with pleiotropic biological activities, has poor water solubility and low bioavailability. The objective of this work was to produce a more bioavailable formulation of QR (liposomes) and to determine the effect of its intraperitoneal pretreatment on the amelioration of Co-Amox-induced liver damage in male rats. Four groups of rats were defined: control, QR liposomes (QR-lipo), Co-Amox, and Co-Amox and QR-lipo. Liver injury severity in rats was evaluated for all groups through measurement of serum liver enzymes, liver antioxidant status, proinflammatory mediators, and microbiota modulation. The results revealed that QR-lipo reduced the severity of Co-Amox-induced hepatic damage in rats, as indicated by a reduction in serum liver enzymes and total liver antioxidant capacity. In addition, QR-lipo upregulated antioxidant transcription factors SIRT1 and Nrf2 and downregulated liver proinflammatory signatures, including IL-6, IL-1β, TNF-α, NF-κB, and iNOS, with upregulation in the anti-inflammatory one, IL10. QR-lipo also prevented Co-Amox-induced gut dysbiosis by favoring the colonization of Lactobacillus, Bifidobacterium, and Bacteroides over Clostridium and Enterobacteriaceae. These results suggested that QR-lipo ameliorates Co-Amox-induced liver damage by targeting SIRT1/Nrf2/NF-κB and modulating the microbiota.
Collapse
Affiliation(s)
- Mahran Mohamed Abd El-Emam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt;
| | - Mahmoud Mostafa
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt;
| | - Amina A. Farag
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Banha 13518, Egypt;
| | - Heba S. Youssef
- Department of Physiology, Faculty of Medicine, Benha University, Benha 13518, Egypt;
| | - Azza S. El-Demerdash
- Laboratory of Biotechnology, Department of Microbiology, Agriculture Research Centre (ARC), Animal Health Research Institute (AHRI), Zagazig 44516, Egypt;
| | - Heba Bayoumi
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha 13518, Egypt;
| | - Mohammed A. Gebba
- Department of Anatomy and Embryology, Faculty of Medicine, Benha University, Benha 13518, Egypt;
- Department of Anatomy and Embryology, Faculty of Medicine, Merit University, Sohag 82524, Egypt
| | - Sawsan M. El-Halawani
- Department of Biotechnology, Urology and Nephrology Center, Mansoura University, Mansoura 35516, Egypt;
| | - Abdulrahman M. Saleh
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt;
| | - Amira M. Badr
- Pharmacology and Toxicology Department, Faculty of Pharmacy, King Saud University, Riyadh P.O. Box 11451, Saudi Arabia
| | - Shorouk El Sayed
- Department of Microbiology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt;
| |
Collapse
|
41
|
Lu MH, Ding KN, Liang SS, Guo YN, He YM, Tang LP. Resveratrol inhibits oxidative damage in lungs of heat-stressed broilers by activating Nrf2 signaling pathway and autophagy. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 258:114949. [PMID: 37121077 DOI: 10.1016/j.ecoenv.2023.114949] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/28/2023] [Accepted: 04/21/2023] [Indexed: 05/22/2023]
Abstract
The purpose of this study was to investigate the effects of resveratrol on heat stress-induced lung injury in broilers and the mechanism underlying this process. Sixty two-week-old SPF BWEL broilers were randomly divided into the heat stress group (HS), resveratrol group (heat stress + 400 mg/kg resveratrol), and the control group after one week of feeding, with 20 chickens in each group. Broilers in the control group were reared at 23 ± 2 ℃. Those in the HS and resveratrol group were reared under heat stress (35 ℃ ± 2 ℃) for 8 h/day for seven days. Broilers in the resveratrol group were fed a diet supplemented with 400 mg/kg resveratrol two days before the start of the experiment. The feeding was continued for nine days. The results showed that HS decreased body weight (BW), average daily feed intake (ADFI), average daily gain (ADG), and lung weight. It, however, increased the lung index, induced lung congestion, and promoted infiltration of inflammatory cells to the lung. Resveratrol improved growth performance and inhibited heat stress-induced lung damage. Compared with broilers in the control group, the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), NAD(P)H quinone oxidoreductase 1 (NQO1), heme oxygenase-1 (HO-1), Beclin-1, LC3 Ⅰ, and LC3 Ⅱ genes in the lung of heat-stressed broilers was significantly lower. The levels of kelch-like ECH-associated protein 1 (Keap1), NQO1, and HO-1 showed a similar trend with gene expressions. Immunofluorescence indicated that HS inhibited the expression of Nrf2 and LC3B proteins. Finally, the ratio of LC3 Ⅱ/LC3 Ⅰ was also significantly lower in the HS group. Further analyses revealed that resveratrol supplements in feeds enhanced antioxidation in the lung by activating the Nrf2 signaling pathway and autophagy. In conclusion, HS causes oxidative damage and inhibits autophagy in broilers. However, resveratrol protects against lung injury by alleviating oxidative stress and enhancing autophagy.
Collapse
Affiliation(s)
- Meng-Han Lu
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Kang-Ning Ding
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Shao-Shan Liang
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yan-Na Guo
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Yong-Ming He
- School of Life Science and Engineering, Foshan University, Foshan, China.
| | - Lu-Ping Tang
- School of Life Science and Engineering, Foshan University, Foshan, China.
| |
Collapse
|
42
|
Xiao L, Li L, Huang J, Luan Y, Pan J, Gai Y, Xu Z. Salidroside attenuates lipopolysaccharide-induced neuroinflammation and cognitive impairment in septic encephalopathy mice. Int Immunopharmacol 2023; 117:109975. [PMID: 36948107 DOI: 10.1016/j.intimp.2023.109975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/13/2023] [Accepted: 02/28/2023] [Indexed: 03/24/2023]
Abstract
Salidroside (SAL) is a natural bioactive compound with anti-oxidative, anti-inflammatory, and neuroprotective properties. In the present study, we generate an experimental design to investigate SAL-mediated protective effect and underlying mechanism on lipopolysaccharide (LPS)-induced neuroinflammation and cognitive impairment in the septic encephalopathy mice model (SEMM). In SEMM, Open-Field Test (OFT) and Novel Object Recognition Test evaluated LPS-induced cognitive impairment, behavioural phenotypes, and memory impairment (NOR). Cytokines and protein expression were assessed using ELISA assay, RT-qPCR, and Western blotting. Our results showed cognitive dysfunction could be reversed when treated with SAL in SEMM. SAL treatment significantly reduced apoptotic TUNEL-positive cells and related gene expression (BAX and BCL-2) and considerably improved neuronal damage in SEMM. In addition, it markedly reduced the production of inflammatory cytokines (TNF-α, IL-1β, and IL-6) and Iba-1-positive cells responsible for microglial activation in mice hippocampus (P < 0.05). The effects of SAL on ROS and oxidative stress markedly reduced malondialdehyde (MDA) content and increased superoxide dismutase (SOD) and catalase (CAT) in the hippocampal tissues of mice. Besides, SAL treatment enhanced LPS-induced autophagy in mice's hippocampus and increased autophagy-related protein expression (Beclin-1 and P62). In addition, the NLRP3 inflammasome pathway and its related proteins (NLRP3, ASC, and cleaved caspase-1) were suppressed by SAL treatment. However, SAL activated the SIRT1/Nrf2 pathway and exerts protection by enhanced expression of the proteins (SIRT1 and Nrf2) and downstream genes (HO-1 and NQO1). Our finding demonstrated that SAL employed neuroprotective effects in SEMM by promoting autophagy via activation of the SIRT1 pathway.
Collapse
Affiliation(s)
- Lingling Xiao
- Department of Emergency Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Pudong New Area, Shanghai 200137, China
| | - Liang Li
- Department of Emergency Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Pudong New Area, Shanghai 200137, China
| | - Jingcong Huang
- Department of Emergency Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Pudong New Area, Shanghai 200137, China
| | - Yuting Luan
- Department of Emergency Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Pudong New Area, Shanghai 200137, China
| | - Jiaying Pan
- Department of Emergency Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Pudong New Area, Shanghai 200137, China
| | - Yun Gai
- General Practice Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Pudong New Area, Shanghai 200137, China.
| | - Zhenyu Xu
- Department of Emergency Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Pudong New Area, Shanghai 200137, China.
| |
Collapse
|
43
|
Xie M, Wang H, Gao T, Peng J, Meng P, Zhang X, Guo D, Liu G, Shi J, Peng Q. The protective effect of luteolin on the depression-related dry eye disorder through Sirt1/NF-κB/NLRP3 pathway. Aging (Albany NY) 2023; 15:261-275. [PMID: 36641776 PMCID: PMC9876631 DOI: 10.18632/aging.204479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/29/2022] [Indexed: 01/16/2023]
Abstract
Luteolin has been reported to exhibit therapeutic effect on depressive-like behaviors in mice. Nevertheless, the therapeutic effect of luteolin on the depression-related dry eye disorder remains inconclusive. In this study, C57 mice were subjected to chronic unpredictable mild stress in a dry environment (relative humidity in the cage <40%). The behavioral test and phenol red cotton thread test were employed to select the mice with both dry eye and depression-like behavior. The mechanism of luteolin on depression-related dry eye disorder was assessed by the Sirt1 selective inhibitor EX-527. Luteolin alleviated depressive-like behaviors induced by CUMS, increased tear secretion and restored corneal defects in mice. The secretions of pro-inflammatory factors IL-1β, IL-6, IL-18 and TNF-α were decreased in hippocampi and corneal tissues by Luteolin treatment. Luteolin treatment up-regulated Sirt1 expression and down-regulated Ac-NF-κB, NLRP3, Ac-Caspase-1, GSDMD-N, Cleaved IL-1β, and Cleaved IL-18 expressions. In addition, the selective inhibition of Sirt1 could weaken the therapeutic effect of luteolin on depression-related dry eye disorder. The beneficial effect of luteolin through Sirt1/NF-κB/NLRP3 signaling pathway might be a therapeutic strategy for the depression-related dry eye disorder.
Collapse
Affiliation(s)
- Mingxia Xie
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- National Key Laboratory Cultivation Base of Chinese Medicinal Powder and Innovative Medicinal Jointly Established by Province and Ministry, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Hanqing Wang
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Tiantian Gao
- College of Pharmacy, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jun Peng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Pan Meng
- National Key Laboratory Cultivation Base of Chinese Medicinal Powder and Innovative Medicinal Jointly Established by Province and Ministry, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Xi Zhang
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Dongwei Guo
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Guangya Liu
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Jian Shi
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Qinghua Peng
- College of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| |
Collapse
|
44
|
Taatjes DJ, Roth J. In focus in HCB. Histochem Cell Biol 2023; 159:1-3. [PMID: 36622387 PMCID: PMC9827435 DOI: 10.1007/s00418-022-02176-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- Douglas J. Taatjes
- grid.59062.380000 0004 1936 7689Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT 05405 USA
| | - Jürgen Roth
- grid.7400.30000 0004 1937 0650University of Zurich, CH-8091 Zurich, Switzerland
| |
Collapse
|
45
|
Liu Y, Guo X, Yu L, Huang Y, Guo C, Li S, Yang X, Zhang Z. Luteolin alleviates inorganic mercury-induced liver injury in quails by resisting oxidative stress and promoting mercury ion excretion. Mol Biol Rep 2023; 50:399-408. [PMID: 36336778 DOI: 10.1007/s11033-022-08049-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 10/19/2022] [Indexed: 11/08/2022]
Abstract
BackgroundInorganic mercury is a well-known toxic substance that can cause oxidative stress and liver damage. Luteolin (Lut) is a kind of natural antioxidant, which is widely found in plants. Therefore, we focused on exploring the alleviative effect of Lut on liver injury induced by mercuric chloride (HgCl2), and the potential molecular mechanism of eliminating mercury ions in quails.Methods and resultsTwenty-one-day-old male quails were randomly split into four groups: control group, Lut group, HgCl2 group, and HgCl2 + Lut group. The test period was 12 weeks. The results showed that Lut could significantly ameliorate oxidative stress, the release of inflammatory factors, and liver damage caused by HgCl2, and reduce the accumulation of Hg2+ in quail liver. Furthermore, Lut evidently increased the levels of protein kinase C α (PKCα), nuclear factor-erythroid-2-related factor 2 (Nrf2), and its downstream proteins, and inhibited nuclear factor-kappaB (NF-κB) production in the liver of quails treated by HgCl2.ConclusionsTo sum up, our results suggest that Lut not only reduces the levels of oxidative stress and inflammation, but also promotes the excretion of Hg2+ by promoting the PKCα/Nrf2 signaling pathway to alleviate HgCl2-induced liver injury in quails.
Collapse
Affiliation(s)
- Yan Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China.,College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, 028000, China
| | - Xinyu Guo
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Lu Yu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Yuxiang Huang
- Branch of Animal Husbandry and Veterinary of Heilongjiang Academy of Agricultural Sciences, Qiqihar, 161000, China
| | - Changming Guo
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Xu Yang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin, 150030, China.
| |
Collapse
|
46
|
Zannino L, Pagano A, Casali C, Oldani M, Balestrazzi A, Biggiogera M. Mercury chloride alters heterochromatin domain organization and nucleolar activity in mouse liver. Histochem Cell Biol 2023; 159:61-76. [PMID: 36136163 PMCID: PMC9899742 DOI: 10.1007/s00418-022-02151-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2022] [Indexed: 02/07/2023]
Abstract
Mercury is a highly toxic element that induces severe alterations and a broad range of adverse effects on health. Its exposure is a global concern because it is widespread in the environment due to its multiple industrial, domestic, agricultural and medical usages. Among its various chemical forms, both humans and animals are mainly exposed to mercury chloride (HgCl2), methylmercury and elemental mercury. HgCl2 is metabolized primarily in the liver. We analysed the effects on the nuclear architecture of an increasing dosage of HgCl2 in mouse hepatocytes cell culture and in mouse liver, focusing specifically on the organization, on some epigenetic features of the heterochromatin domains and on the nucleolar morphology and activity. Through the combination of molecular and imaging approaches both at optical and electron microscopy, we show that mercury chloride induces modifications of the heterochromatin domains and a decrease of some histones post-translational modifications associated to heterochromatin. This is accompanied by an increase in nucleolar activity which is reflected by bigger nucleoli. We hypothesized that heterochromatin decondensation and nucleolar activation following mercury chloride exposure could be functional to express proteins necessary to counteract the harmful stimulus and reach a new equilibrium.
Collapse
Affiliation(s)
- Lorena Zannino
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, 27100 Pavia, Italy
| | - Andrea Pagano
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, 27100 Pavia, Italy
| | - Claudio Casali
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, 27100 Pavia, Italy
| | - Monica Oldani
- Department of Biology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy
| | - Alma Balestrazzi
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, 27100 Pavia, Italy
| | - Marco Biggiogera
- Department of Biology and Biotechnology ‘L. Spallanzani’, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
47
|
The Ethanolic Extract of Lindera aggregata Modulates Gut Microbiota Dysbiosis and Alleviates Ethanol-Induced Acute Liver Inflammation and Oxidative Stress SIRT1/Nrf2/NF- κB Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6256450. [PMID: 36583098 PMCID: PMC9794438 DOI: 10.1155/2022/6256450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/10/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
This study is an attempt to evaluate the therapeutic effect of the ethanolic extract of Lindera aggregata on the liver and intestinal microbiota in rats with alcohol-induced liver injury (ALI). Rats were treated with 70 mg probiotics, 1 g/kg, 2 g/kg, and 3 g/kg ethanolic extract of Lindera aggregata, respectively, for 10 days. We found that Lindera aggregata could significantly reduce the biochemical parameters in the serum of ALD rats. Lindera aggregata alleviates oxidative stress and inflammation by upregulating SIRT1 and Nrf2 and downregulating COX2 and NF-κB. The results of 16S rRNA gene sequencing showed that the medium dose of Lindera aggregata had the best effect on the growth of beneficial bacteria. Diversity analysis and LEfSe analysis showed that beneficial bacteria gradually occupied the dominant niche. The relative abundance of potential pathogens in the gut decreased significantly. We demonstrated that the ethanolic extract of Lindera aggregata can alleviate the oxidative stress and inflammation induced by alcohol through the SIRT1/Nrf2/NF-κB pathway and can modulate the disturbance of gut microbiota induced by alcohol intake.
Collapse
|
48
|
Jiang H, Mao T, Sun Z, Shi L, Han X, Zhang Y, Zhang X, Wang J, Hu J, Zhang L, Li J, Han H. Yinchen Linggui Zhugan decoction ameliorates high fat diet-induced nonalcoholic fatty liver disease by modulation of SIRT1/Nrf2 signaling pathway and gut microbiota. Front Microbiol 2022; 13:1001778. [PMID: 36578580 PMCID: PMC9791106 DOI: 10.3389/fmicb.2022.1001778] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
Yinchen Linggui Zhugan decoction (YLZD) is an effective and classical traditional herbal prescription for treating the nonalcoholic fatty liver disease (NAFLD) and has been proven to be effective in the regulation of lipid metabolism disorder and attenuate inflammation for a NAFLD rat model. However, the exact underlying mechanism has not been elucidated. In the current study, a NAFLD rat model was established using a high-fat diet (HFD) for 10 weeks, followed by YLZD treatment with 1.92 g/kg/day for 4 weeks to explore the mechanisms of YLZD. Our results showed that YLZD decreased the hepatic lipid deposition, restored the liver tissue pathological lesions, inhibited the expression of oxidative stress, and decreased the inflammatory cytokines levels. Meanwhile, the genes and proteins expressions of SIRT1/Nrf2 signaling pathway together with downstream factors including HO-1 and NQO1 were elevated in the YLZD treated NAFLD rats. For further elaborating the upstream mechanism, short-chain fatty acids (SCFAs) in serum and feces were measured by liquid chromatograph mass spectrometer and gas chromatograph mass spectrometer, and the differences in gut microbiota of rats in each group were analyzed through high-throughput sequencing of 16S rRNA. The results demonstrated that the contents of butyric acid (BA) and total SCFAs in YLZD-treated NAFLD rats were significantly increased in serum and feces. 16S rRNA sequencing analysis illustrated that YLZD intervention led to a modification of the gut microbiota composition, with a decrease of Oribacterium, Lactobacillus and the ratio of Firmicutes/Bacteroides, as well as the increase in SCFAs-producing bacteria such as Christensenellaceae, Clostridia, Muribaculaceae, and Prevotellaceae. Spearman rank correlation analysis indicated that BA and total SCFAs were negatively co-related with oxidative stress-related factors and inflammatory cytokines, while they were positively co-related with SIRT1/Nrf2 pathway related genes and proteins. Furthermore, in vitro study confirmed that BA effectively reduced oxidative stress by activating SIRT1/Nrf2 signaling pathway in L02 cells. Together, the present data revealed YLZD could ameliorate HFD-induced NAFLD in rats by the modulation of SIRT1/Nrf2 signaling pathway and gut microbiota.
Collapse
Affiliation(s)
- Hui Jiang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhongmei Sun
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Shi
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Han
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Zhang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaosi Zhang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiali Wang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Juncong Hu
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Liming Zhang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China,*Correspondence: Junxiang Li, Haixiao Han
| | - Haixiao Han
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China,*Correspondence: Junxiang Li, Haixiao Han
| |
Collapse
|
49
|
Deng N, Lv Y, Bing Q, Li S, Han B, Jiang H, Yang Q, Wang X, Wu P, Liu Y, Zhang Z. Inhibition of the Nrf2 signaling pathway involved in imidacloprid-induced liver fibrosis in Coturnix japonica. ENVIRONMENTAL TOXICOLOGY 2022; 37:2354-2365. [PMID: 35716027 DOI: 10.1002/tox.23601] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 06/15/2023]
Abstract
Imidacloprid (IMI) is a kind of widely used neonicotinoid insecticide. However, the toxicity of IMI is not only applied to target pests but also causes serious negative effects on birds and other creatures. Our previous studies have shown that long-term exposure to IMI can induce liver fibrosis in quails. However, the specific mechanism of quail liver fibrosis induced by IMI is not completely clear. Accordingly, the purpose of this study is to further clarify the potential molecular mechanism of IMI-induced liver fibrosis in quails. Japanese quails (Coturnix japonica) were treated with/without IMI (intragastric administration with 6 mg/kg body weight) in the presence/absence of luteolin (Lut) (fed with 800 mg/kg) for 90 days. The results reveal that IMI can induce hepatic fibrosis, oxidative stress, fatty degeneration, inflammation, and the down-expression of nuclear factor-E2-related factor-2 (Nrf2). Furthermore, the treatment of Lut, a kind of Nrf2 activator, increased the expression of Nrf2 in livers and alleviated liver fibrosis in quails. Altogether, our study demonstrates that inhibition of the Nrf2 pathway is the key to liver fibrosis induced by IMI in quails. These results provide a new understanding for the study of the toxicity of IMI and a practical basis for the treatment of liver fibrosis caused by IMI.
Collapse
Affiliation(s)
- Ning Deng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yueying Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
- Department of Laboratory Animal Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qizheng Bing
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Bing Han
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Huijie Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Qingyue Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiaoqiao Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Pengfei Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yan Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
50
|
Wang H, Yao X, Huang K, Zhang J, Xiao J, Guo J, Wei D, Xiang B. Low-dose dexamethasone in combination with luteolin improves myocardial infarction recovery by activating the antioxidative response. Biomed Pharmacother 2022; 151:113121. [PMID: 35605295 DOI: 10.1016/j.biopha.2022.113121] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 11/15/2022] Open
Abstract
This study aimed to explore the effects of dexamethasone (DEX) and its combination with luteolin (LUT) on cardiac function during myocardial infarction (MI) in a mouse model. We evaluated whether the Keap1/Nrf2 pathway mediates the cardioprotective function of DEX both in vivo and in vitro. The MI mouse model was established by ligation of the left anterior descending coronary artery of wild-type (WT) and Nrf2 knockout mice. After recovery for 21 days, DEX or its combination with LUT was intraperitoneally administered at different doses to WT or Nrf2 knockout mice daily for 7 consecutive days. Mice treated with DEX at a low dose (50 μg/kg/day) showed better cardiac function, fewer cardiac lesions, and smaller infarct sizes compared with MI model mice. DEX (50 μg/kg/day) administration also significantly decreased the production of reactive oxygen species (ROS) and pro-inflammatory cytokines, increased the expression of antioxidative enzymes, and activated the Keap1/Nrf2/HO-1 pathway. However, in Nrf2 knockout mice, DEX treatment did not influence cardiac function, inflammation, the oxidative response, or Keap1/Nrf2/HO-1 activation. In the MI cell model, low concentrations of DEX attenuated the H2O2-induced decreases in cell viability and antioxidative enzyme levels and activated the Keap1/Nrf2/HO-1 pathway. Low doses of DEX exerted protective effects in MIR mice and MI cell models by improving cardiac function, eliminating ROS, inhibiting inflammatory responses, and activating antioxidative responses. The protective effects of DEX on myocardial tissues were mediated by the Keap1/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Xiaoqin Yao
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Keli Huang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Jing Zhang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Jingrong Xiao
- Nursing Department, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Jing Guo
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Dachuang Wei
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Bo Xiang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| |
Collapse
|