1
|
Salem GA, Aref M, El-Malkey NF, Alqahtani HA, Abd-Almotaleb NA, Nassan MA, Elsherbiny H. Exercise induced irisin mitigates hepatitis in anabolic-androgenic steroids treated rats via modulation of PGC-1-α/PPARγ/Nrf2 and NRF2/NF-κB/TLR4 signaling. Tissue Cell 2025; 95:102829. [PMID: 40054305 DOI: 10.1016/j.tice.2025.102829] [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: 12/02/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 05/15/2025]
Abstract
Irisin, a myokine released during exercise, has been shown to exert protective effects against metabolic and inflammatory disorders. Its role in mitigating hepatic damage induced by anabolic-androgenic steroids (AAS) remains largely unexplored. This study was conducted to examine the effects of exercise on irisin level and its capability to prevent hepatotoxicity caused by anabolic androgenic steroids (AAS) in rat model. The fifty-two male rats were divided into four groups: control, AAS treated (15 mg/kg/day S.C/8 W), exercised, and exercised- AAS treated. The following procedures were carried out: liver function tests, serum irisin, tissue inflammatory and oxidative stress markers, macro and micromorphological evaluation, and the examination of nuclear factor erythroid 2-related factor 2 (Nrf2), peroxisome proliferator-activated receptor-gamma (PPARγ) and its coactivator-1α (PGC1α) by immunohistochemistry. The liver tissue's expression of nuclear factor kappa B (NF-κB), Toll-like receptor-4 (TLR4), and Nrf2 mRNA was also assessed. After administering AAS to animals, aerobic exercise was found to significantly improve liver function tests, inflammation, and oxidative stress, reduce liver weight, improve morphological and histological changes, and improve the hepatic injury score. Furthermore, there was a notable rise in serum irisin, hepatic PPARγ, PGC1α, and Nrf2 immune-expressions and Nrf2 mRNA expression, while NF-κB and TLR4 mRNA expressions were significantly decreased. In conclusion, the irisin/PGC1α/PPARγ/Nrf2 and Nrf2/NF-κB/TLR4 signaling pathways may be modulated by aerobic exercise, which also reduces the liver's oxidative stress and inflammatory reactions to AAS treatment.
Collapse
Affiliation(s)
- Gamal A Salem
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, El-Sharkia 44519, Egypt.
| | - Mohamed Aref
- Department of Anatomy and embryology, Faculty of Veterinary medicine, Zagazig University, El-Sharkia 44519, Egypt.
| | - Nanees F El-Malkey
- Department of Medical physiology, Faculty of medicine, Zagazig University, Zagazig, El-Sharkia 44519, Egypt
| | - Haifa A Alqahtani
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Noha Ali Abd-Almotaleb
- Department of Medical Anatomy, Faculty of Medicine, Zagazig University, El-Sharkia 44519, Egypt
| | - Mohamed A Nassan
- Department of clinical laboratory sciences, Turabah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Hadeel Elsherbiny
- Department of Medical physiology, Faculty of medicine, Zagazig University, Zagazig, El-Sharkia 44519, Egypt
| |
Collapse
|
2
|
Hassan NH, Kamel GM, Fayed HM, Korany RMS, Ramadan A. Dapagliflozin alleviates thioacetamide induced-liver fibrosis in rats via controlling the Nrf2/HO-1 and TLR4/TGF-β1/PI3K signaling pathways. Immunopharmacol Immunotoxicol 2025; 47:392-405. [PMID: 40296648 DOI: 10.1080/08923973.2025.2496661] [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: 01/23/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVES Because liver fibrosis causes several insults that can result in death, it is regarded as an epidemic health issue. As "an inhibitor of the sodium-glucose cotransporter-2 (SGLT2)," Dapagliflozin (Dapa) is one of the newest anti-diabetic drugs used to treat type 2 diabetes mellitus. Dapa's antioxidant, anti-inflammatory, and antifibrotic properties produced positive impacts in numerous human and animal models. Due to Dapa's previously documented properties, we planned this investigation to elucidate the protective function of Dapa in male rat liver fibrosis caused by thioacetamide (TAA) as well as the expected pathways. METHODS There were four groups of 24 rats: a control group, a TAA group that received (100 mg/kg b.wt intraperitoneally twice a week for 6 weeks), "TAA + Dapa" groups that given oral Dapa at (1 and 2 mg/kg b.wt. for 4 weeks in addition to TAA injections). RESULTS It was shown that TAA injections increased toll-like receptor 4 (TLR4) (509.6%), tumor necrosis factor (TNF-α) (298.8%), alanine aminotransferase (ALT), aspartate aminotransferase (AST), malondialdehyde (MDA), interleukin-6 (IL-6) (330.9%), phosphotidylinositol-3-kinase (PI3K) (428.9% %), and transforming growth factor-beta (TGF-β1) (416.6%) levels. All of these markers were considerably reduced by Dapa treatment. In addition, reduced glutathione (GSH), nuclear factor erythroid 2-related factor 2 (Nrf2) (79%), albumin, Heme-oxygenase 1 (HO-1) (69%), and superoxide dismutase (SOD) were all decreased after TAA injection; however, they were restored by Dapa administration. The Dapa-treated groups had higher Nrf2 and HO-1 gene expressions, based on the results of PCR. Biochemical outcomes were validated by histopathological results. Immunohistopathological study revealed that DAPA treatment decreased caspase-3 and alpha-smooth Muscle Actin (αSMA) expression. CONCLUSION Due to its interactions with the Nrf2/HO-1 and TLR4 pathways, our research showed that Dapa had antioxidant and anti-inflammatory qualities against TAA-induced liver fibrosis.
Collapse
Affiliation(s)
- Nourhan Hussien Hassan
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Gehan M Kamel
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hany M Fayed
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Reda M S Korany
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Amer Ramadan
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
3
|
Jayaraman S, Prasad M, Natarajan SR, Krishnamoorthy R, Alshuniaber MA, Gatasheh MK, Veeraraghavan VP, Rajagopal P, Palanisamy CP. Molecular mechanisms underlying the effects of beta-sitosterol on TGF-β1/Nrf2/SIRT1/p53-mediated signaling in the kidney of a high-fat diet and sucrose-induced type-2 diabetic rat. Chem Biol Interact 2025; 411:111443. [PMID: 39986364 DOI: 10.1016/j.cbi.2025.111443] [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: 11/16/2024] [Revised: 01/23/2025] [Accepted: 02/20/2025] [Indexed: 02/24/2025]
Abstract
Diabetic nephropathy, a severe problem of diabetes mellitus, is exacerbated by high-fat diets, prompting a need for interventions. Previous study from our laboratory has shown that β-sitosterol, a potent plant sterol has anti-inflammatory and glucose-lowering efficacy by involving insulin metabolic signalling pathway but its role on anti-oxidant signaling pathways, play a crucial role in mitigating oxidative stress and inflammation associated diabetic nephropathy, highlighting its importance as a potential therapeutic target for managing this debilitating complication of diabetes is unknown. This study was aimed to intricate the molecular mechanisms involved in the potential of β-sitosterol (BSIT) on TGF-β1/Nrf2/SIRT1/p53 signaling in high fat diet (HFD) and sucrose induced diabetic nephropathy (DN) in the rat kidney by employing various comprehensive bioinformatic analysis. We have used various comprehensive methods such as pathway predictions, Drug-Protein Interaction, Functional annotation analysis, and molecular docking techniques. Further, in vivo analysis of BSIT on biochemical profiles, gene and protein expression analysis of anti-oxidant and inflammatory signaling molecules was performed in the kidney of high fat diet (HFD) and sucrose-induced diabetic nephropathy. Computational studies provided insights into β-sitosterol's binding affinities and interaction modes with key proteins, suggesting its potential to regulate TGF-β1/Nrf2/SIRT1/p53 signaling pathways. Results of in vivo findings validated computational predictions, showcasing BSIT's multifaceted effects in mitigating diabetic nephropathy and associated complications including regulation of lipid metabolism, combating oxidative stress, and inflammation. The findings underscore BSIT's therapeutic potential by preserving cellular viability, regulating cell death, enhancing antioxidant defence, and stabilizing metabolic processes. Our study concludes that BSIT's ability to potentially regulate TGF-β1/Nrf2/SIRT1/p53 pathways, emphasizing its promising role in managing diabetic nephropathy and associated complications.
Collapse
MESH Headings
- Animals
- Sirtuin 1/metabolism
- NF-E2-Related Factor 2/metabolism
- Sitosterols/pharmacology
- Sitosterols/therapeutic use
- Sitosterols/metabolism
- Sitosterols/chemistry
- Diet, High-Fat/adverse effects
- Signal Transduction/drug effects
- Tumor Suppressor Protein p53/metabolism
- Rats
- Transforming Growth Factor beta1/metabolism
- Male
- Kidney/metabolism
- Kidney/drug effects
- Kidney/pathology
- Sucrose
- Molecular Docking Simulation
- Diabetic Nephropathies/metabolism
- Diabetic Nephropathies/drug therapy
- Diabetic Nephropathies/pathology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/drug therapy
- Oxidative Stress/drug effects
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, India.
| | - Monisha Prasad
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, India.
| | - Sathan Raj Natarajan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, India.
| | - Rajapandiyan Krishnamoorthy
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Mohammad A Alshuniaber
- Department of Food Science and Nutrition, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Mansour K Gatasheh
- Department of Biochemistry, College of Science, King Saud University, P.O.Box 2455, Riyadh, 11451, Saudi Arabia.
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, India.
| | - Ponnulakshmi Rajagopal
- Central Research Laboratory, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research (Deemed to be University), Chennai, 600095, India.
| | - Chella Perumal Palanisamy
- Department of Chemical Technology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
4
|
Shaban DA, El-Shahawy AAG, Zanaty MI, Eldin ZE, Abd-Elbaset M, Shams A, Tamur S, Ahmed OM. Utilizing Nanoparticles of Hesperidin Loaded on Layered Double Hydroxide to Reduce Hepatotoxicity Caused by Paracetamol in Rats: Controlling of Biotransformation, Oxidative Stress, Inflammation, and Apoptosis. Pharmaceutics 2025; 17:429. [PMID: 40284423 PMCID: PMC12030007 DOI: 10.3390/pharmaceutics17040429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: The most used antipyretic and pain relief treatment is paracetamol (acetaminophen), also known as N-acetyl-para-aminophenol (APAP). However, it is considered potentially hazardous if consumed repeatedly in large doses or over prolonged periods. This investigation explores the effectiveness of hesperidin (Hesp) and Hesp loaded on layered double hydroxide nanoparticles (Hesp-NPs) in inhibiting the progression of acute hepatotoxicity in rats induced by APAP. Methods: LDH-Hesp-NPs were prepared and characterized. Male Wistar rats were orally treated with Hesp and Hesp-NPs at the same adjusted dose (100 mg/kg) every other day for six weeks. After 2 h of the first doses of Hesp and Hesp-NPs, the rats received one oral dose of APAP (750 mg/kg). Results: Administering of Hesp and Hesp-NPs to APAP-treated rats significantly reduced oxidant parameter (malondialdehyde) and serum enzymes (ALT, AST, LDH, and ALP) associated with liver function. Antioxidant markers in the liver, such as catalase and glutathione, also increased notably. Moreover, Hesp and Hesp-NPs enhanced the mRNA expression of liver UGT1A6, IL-10, and HO-1. Conversely, the mRNA expressions of liver CYP1A1, KEAP1, TGF-β, P53, and BAX decreased. These improvements in biochemical and molecular markers were corroborated by liver histopathology. Conclusions: Hesp and Hesp-NPs protect significantly against APAP-induced hepatotoxicity in male Wistar rats. Hesp-NPs treatment was more potent. The protective effects may be mediated via modulation of APAP biotransformation, oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Deyaa A. Shaban
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt; (D.A.S.); (M.I.Z.)
| | - Ahmed A. G. El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt; (A.A.G.E.-S.); (Z.E.E.)
| | - Mohamed I. Zanaty
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt; (D.A.S.); (M.I.Z.)
| | - Zienab E. Eldin
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt; (A.A.G.E.-S.); (Z.E.E.)
| | - Mohamed Abd-Elbaset
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, El Salehya El Gadida University (SGU), El Salheya 44813, Egypt
| | - Anwar Shams
- Department of Pharmacology, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Research Center for Health Sciences, Deanship of Graduate Studies and Scientific Research, Taif University, Taif 26432, Saudi Arabia
- High Altitude Research Center, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Shadi Tamur
- Department of Pediatric, College of Medicine, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Osama M. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62521, Egypt;
| |
Collapse
|
5
|
Elazab ST, Hsu WH. α-Bisabolol alleviates doxorubicin-induced cognitive dysfunction in rats via enhancing the hippocampal BDNF/TrKB signaling and inhibiting neuroinflammation. Front Pharmacol 2025; 16:1549009. [PMID: 40124785 PMCID: PMC11925949 DOI: 10.3389/fphar.2025.1549009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Chemofog is a serious sequela commonly manifested among cancer patients receiving doxorubicin (DOX) chemotherapy. Our goal was to explore the abrogative action of α-Bisabolol (BISA), a phytochemical sesquiterpene, against DOX-induced cognitive deficit. Rats were allocated into 5 groups: Group I: control; Group II received BISA orally (100 mg/kg/day for 4 weeks); Group III received DOX (2 mg/kg/week/i.p.) for 4 weeks; Groups IV and V were administered BISA orally at 50 and 100 mg/kg, respectively plus DOX, i. p. Results: 1) BISA attenuated DOX-induced chemofog as shown in memory-related behavioral tests. 2) BISA restored the hippocampal histological structure and redox homeostasis via diminishing MDA content and upregulating Nrf2 and HO-1 genes. 3) BISA mitigated DOX-induced neuroinflammation through reducing NF-kB, TNF-α, IL-6, IL-1β, and GFAP expressions. 4) BISA repressed the hippocampal apoptosis via downregulating Bax gene and upregulating Bcl-2 gene. 5) BISA enhanced the synaptic plasticity by activating the BDNF/TrKB signaling and increasing the levels of neurotransmitters that enhance memory, i.e., ACh, 5-HT, and DA. BISA at 100 mg/kg/day exerted a better neuroprotection than BISA at 50 mg/kg/day. Thus, BISA may protect cancer patients from cognitive disorders caused by DOX.
Collapse
Affiliation(s)
- Sara T. Elazab
- Department of Pharmacology, Facultyof Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Walter H. Hsu
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
6
|
Huchzermeier R, van der Vorst EPC. Aryl hydrocarbon receptor (AHR) and nuclear factor erythroid-derived 2-like 2 (NRF2): An important crosstalk in the gut-liver axis. Biochem Pharmacol 2025; 233:116785. [PMID: 39890034 DOI: 10.1016/j.bcp.2025.116785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/18/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, mainly involved in detoxification. However, in the intestine, metabolites derived from the diet, which are converted by a wide range of bacteria can also activate the AHR. This intestinal AHR activation plays a key role in maintaining the gut barrier by, for example, upregulating antimicrobial peptides and anti-inflammatory cytokines. Since the gut barrier influences the gut-liver axis by regulating the leaking of metabolites, bacteria, and endotoxins into circulation and particularly into the liver, the AHR is a key factor in the gut-liver axis. Vice versa, certain liver pathologies also influence the gut microbiome, thereby altering bacteria-derived activation of the AHR. Additionally, bile acids can impact the gut via the liver and thereby also affect the AHR. The aryl hydrocarbon receptor (AHR) interacts with several molecular factors, one of which is the nuclear factor erythroid-derived 2-like 2 (NRF2), a transcription factor primarily associated with regulating antioxidant stress responses. The interplay between AHR and NRF2 has been investigated in the context of various diseases; this review highlights the significance of this interaction within the framework of the gut-liver axis.
Collapse
Affiliation(s)
- Rosanna Huchzermeier
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany; Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany; Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany.
| |
Collapse
|
7
|
Ashour NA, El-Masry TA, El-Mahdy NA, E Khodier A, Elmorshedy KE, Gaballa MMS, Negm WA. A novel combination therapy using Dapagliflozin and Cycas media extract in experimentally induced diabetic wounds by targeting novel pathways in wound healing. Int Immunopharmacol 2025; 144:113618. [PMID: 39615109 DOI: 10.1016/j.intimp.2024.113618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/02/2024] [Accepted: 11/08/2024] [Indexed: 12/15/2024]
Abstract
Diabetes mellitus, a globally prevalent condition, often complicates wound healing, leading to chronic, non-healing wounds. This study explores a novel combination therapy using Dapagliflozin and Cycas media extract for treating experimentally induced diabetic wounds in rats. By targeting the Notch signaling pathway, a critical pathway in wound healing, this research investigates the efficacy of this combination therapy in accelerating wound repair. Forty-two male Wistar albino rats were divided into control and treatment groups, receiving various Dapagliflozin and Cycas media gel combinations. The study evaluated wound healing, biochemical markers, gene expression, and histopathological changes. The findings suggest that the combination therapy significantly enhances wound healing, modulates oxidative stress, alters inflammatory responses, and influences key genes in the Notch pathway. This research provides a new perspective on diabetic wound management and underlines the potential of combining Dapagliflozin and Cycas media as a therapeutic approach.
Collapse
Affiliation(s)
- Nada A Ashour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Nageh A El-Mahdy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Ahmed E Khodier
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt
| | - Kadreya E Elmorshedy
- Department of Anatomy, Faculty of Medicine, Tanta University, Tanta 31527, Egypt; Department of Anatomy, Faculty of Medicine, King Khaled University, Saudi Arabia
| | - Mohamed M S Gaballa
- Department of Pathology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
8
|
Pan F, Wang CN, Yu ZH, Wu ZR, Wang Z, Lou S, Li WH, Liu GX, Li T, Zhao YZ, Tang Y. NADPHnet: a novel strategy to predict compounds for regulation of NADPH metabolism via network-based methods. Acta Pharmacol Sin 2024; 45:2199-2211. [PMID: 38902503 PMCID: PMC11420228 DOI: 10.1038/s41401-024-01324-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/26/2024] [Indexed: 06/22/2024]
Abstract
Identification of compounds to modulate NADPH metabolism is crucial for understanding complex diseases and developing effective therapies. However, the complex nature of NADPH metabolism poses challenges in achieving this goal. In this study, we proposed a novel strategy named NADPHnet to predict key proteins and drug-target interactions related to NADPH metabolism via network-based methods. Different from traditional approaches only focusing on one single protein, NADPHnet could screen compounds to modulate NADPH metabolism from a comprehensive view. Specifically, NADPHnet identified key proteins involved in regulation of NADPH metabolism using network-based methods, and characterized the impact of natural products on NADPH metabolism using a combined score, NADPH-Score. NADPHnet demonstrated a broader applicability domain and improved accuracy in the external validation set. This approach was further employed along with molecular docking to identify 27 compounds from a natural product library, 6 of which exhibited concentration-dependent changes of cellular NADPH level within 100 μM, with Oxyberberine showing promising effects even at 10 μM. Mechanistic and pathological analyses of Oxyberberine suggest potential novel mechanisms to affect diabetes and cancer. Overall, NADPHnet offers a promising method for prediction of NADPH metabolism modulation and advances drug discovery for complex diseases.
Collapse
Affiliation(s)
- Fei Pan
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Cheng-Nuo Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhuo-Hang Yu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zeng-Rui Wu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Ze Wang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Shang Lou
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wei-Hua Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Gui-Xia Liu
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Ting Li
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Yu-Zheng Zhao
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yun Tang
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
9
|
Almukainzi M, El-Masry TA, Ibrahim HA, Saad HM, El Zahaby EI, Saleh A, El-Nagar MMF. Ameliorative Effect of Chitosan/ Spirulina platensis Ethanolic Extract Nanoformulation against Cyclophosphamide-Induced Ovarian Toxicity: Role of PPAR-γ/Nrf-2/HO-1 and NF-kB/TNF-α Signaling Pathways. Mar Drugs 2024; 22:395. [PMID: 39330276 PMCID: PMC11433581 DOI: 10.3390/md22090395] [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: 06/10/2024] [Revised: 08/14/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Cyclophosphamide (CP) is an anticancer drug that causes infertility disorders. This study was designed to evaluate a nanoformulation of chitosan with an ethanolic extract from Spirulina platensis in terms of its protection against cyclophosphamide-induced ovarian toxicity. Nine groups of female Wistar rats were randomly assigned as follows: 1: control vehicle, 2: chitosan polymer, 3: telmisartan, 4: Spirulina platensis extract, 5: nanoformulation of the Spirulina platensis, and 6: single injection of CP; groups 7, 8, and 9 received the same treatments as those used in groups 3, 4, and 5, respectively, with a single dose of CP (200 mg/kg, I.P). The results displayed that the CP treatment decreased estradiol, progesterone, anti-mullerian hormone, and GSH content, and it downregulated PPAR-γ, Nrf-2, and HO-1 gene expression. In addition, the CP treatment caused an increase in the FSH, LH, and MDA levels. In the same manner, the protein expression of caspase-3, NF-kB, and TNF-α was upregulated in response to the CP treatment, while PPAR-γ was downregulated in comparison with the control. The rats treated with SPNPs exhibited a substantial reduction in the detrimental effects of oxidative stress and inflammation of the ovarian tissue. This study's conclusions showed that SPNPs counteracted the effects of CP, preventing the death of ovarian follicles and restoring the gonadotropin hormone balance and normal ovarian histological appearance.
Collapse
Affiliation(s)
- May Almukainzi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (M.A.); (A.S.)
| | - Thanaa A. El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (T.A.E.-M.); (H.A.I.)
| | - Hanaa A. Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (T.A.E.-M.); (H.A.I.)
| | - Hebatallah M. Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Cairo 51511, Egypt;
| | - Enas I. El Zahaby
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt;
| | - Asmaa Saleh
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia; (M.A.); (A.S.)
| | - Maysa M. F. El-Nagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (T.A.E.-M.); (H.A.I.)
| |
Collapse
|
10
|
Li R, Zhao C, Zhang Y, Huang W, Wang J, Cao G, Cai Z. PM 2.5-induced DNA oxidative stress in A549 cells and regulating mechanisms by GST DNA methylation and Keap1/Nrf2 pathway. Toxicol Mech Methods 2024; 34:517-526. [PMID: 38293967 DOI: 10.1080/15376516.2024.2307967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 01/15/2024] [Indexed: 02/01/2024]
Abstract
Fine particulate matter (PM2.5) increases the risks of lung cancer. Epigenetics provides a new toxicology mechanism for the adverse health effects of PM2.5. However, the regulating mechanisms of PM2.5 exposure on candidate gene DNA methylation changes in the development of lung cancer remain unclear. Abnormal expression of the glutathione S transferase (GST) gene is associated with cancer. However, the relationship between PM2.5 and DNA methylation-mediated GST gene expression is not well understood. In this study, we performed GST DNA methylation analysis and GST-related gene expression in human A549 cells exposed to PM2.5 (0, 50, 100 µg/mL, from Taiyuan, China) for 24 h (n = 4). We found that PM2.5 may cause DNA oxidative damage to cells and the elevation of GSTP1 promotes cell resistance to reactive oxygen species (ROS). The Kelch-1ike ECH-associated protein l (Keap1)/nuclear factor NF-E2-related factor 2 (Nrf2) pathway activates the GSTP1. The decrease in the DNA methylation level of the GSTP1 gene enhances GSTP1 expression. GST DNA methylation is associated with reduced levels of 5-methylcytosine (5mC), DNA methyltransferase 1 (DNMT1), and histone deacetylases 3 (HDAC3). The GSTM1 was not sensitive to PM2.5 stimulation. Our findings suggest that PM2.5 activates GSTP1 to defend PM2.5-induced ROS and 8-hydroxy-deoxyguanosine (8-OHdG) formation through the Keap1/Nrf2 signaling pathway and GSTP1 DNA methylation.
Collapse
Affiliation(s)
- Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan, PR China
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, PR China
| | - Chao Zhao
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, PR China
| | - Yuexia Zhang
- Institute of Environmental Science, Shanxi University, Taiyuan, PR China
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, PR China
| | - Wei Huang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, PR China
| | - Jiayi Wang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, PR China
| | - Guodong Cao
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, PR China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Hong Kong, PR China
| |
Collapse
|
11
|
Mohamed DA, Fouda K, Mabrok HB, El-Shamarka ME, Hamed IM. Sourdough bread as nutritional intervention tool for improvement of cognitive dysfunction in diabetic rats. BMC Nutr 2024; 10:53. [PMID: 38528644 DOI: 10.1186/s40795-024-00861-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: 05/06/2023] [Accepted: 02/27/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND The current research targeted to study the impact of nutritional intervention by two sourdough breads in improvement of cognitive dysfunction in diabetic rats. METHODS Type-2 diabetes was induced in rats by Streptozotocin-Nicotinamide (STZ-NC). Diabetic rats were fed on balanced diet or balanced diet containing 20% of sourdough bread I or II for a month. Lipid profile, oxidative stress, inflammatory markers and cognitive functions were assessed in all rats. Gene expression of brain-derived neurotrophic factor (BDNF) and nuclear respiratory factor 2 (NRF-2) were assessed in hippocampal tissue, while expression of phosphoenol pyruvate carboxy kinase (PEPCK), and glucose transporter 2 (GLUT2) genes were evaluated in hepatic tissue. Chemical composition and fatty acids profile were evaluated in the prepared sourdough bread. RESULTS Sourdough bread II showed higher content of phenolic compounds, fat, fiber and carbohydrates. Fatty acids profile revealed that sourdough bread I was higher in saturated fatty acids (16.08%), while sourdough bread sample II was higher in unsaturated fatty acids (79.33%). Sourdough bread I or II feeding rats' showed significant improvement in hyperglycemia, oxidative stress markers, inflammatory markers, lipid profile, liver and kidney functions in association with improvement in cognitive function. Gene expression of BDNF and NRF2 in hippocampal tissue were increased significantly, while hepatic GLUT2 and PEPCK gene expression were down-regulated in diabetic given sourdough bread I or II. CONCLUSION Sourdough bread II was superior in all the studied parameters. The anti-diabetic effect and protection from cognitive dysfunction of sourdough bread samples may be ascribed to the occurrence of dietary fibers, phenolic compounds, and polyunsaturated fatty acids.
Collapse
Affiliation(s)
- Doha A Mohamed
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo, 12622, Egypt.
| | - Karem Fouda
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Hoda B Mabrok
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Marwa E El-Shamarka
- Toxicology and Narcotics Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Ibrahim M Hamed
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| |
Collapse
|
12
|
Wakai E, Shiromizu T, Otaki S, Koiwa J, Tamaru S, Nishimura Y. Lansoprazole Ameliorates Isoniazid-Induced Liver Injury. Pharmaceuticals (Basel) 2024; 17:82. [PMID: 38256915 PMCID: PMC10821343 DOI: 10.3390/ph17010082] [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: 11/16/2023] [Revised: 12/22/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Isoniazid is a first-line drug in antitubercular therapy. Isoniazid is one of the most commonly used drugs that can cause liver injury or acute liver failure, leading to death or emergency liver transplantation. Therapeutic approaches for the prevention of isoniazid-induced liver injury are yet to be established. In this study, we identified the gene expression signature for isoniazid-induced liver injury using a public transcriptome dataset, focusing on the differences in susceptibility to isoniazid in various mouse strains. We predicted that lansoprazole is a potentially protective drug against isoniazid-induced liver injury using connectivity mapping and an adverse event reporting system. We confirmed the protective effects of lansoprazole against isoniazid-induced liver injury using zebrafish and patients' electronic health records. These results suggest that lansoprazole can ameliorate isoniazid-induced liver injury. The integrative approach used in this study may be applied to identify novel functions of clinical drugs, leading to drug repositioning.
Collapse
Affiliation(s)
- Eri Wakai
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan; (E.W.); (T.S.); (S.O.); (J.K.)
| | - Takashi Shiromizu
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan; (E.W.); (T.S.); (S.O.); (J.K.)
- Mie University Research Center for Cilia and Diseases, Tsu 514-8507, Mie, Japan
| | - Shota Otaki
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan; (E.W.); (T.S.); (S.O.); (J.K.)
| | - Junko Koiwa
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan; (E.W.); (T.S.); (S.O.); (J.K.)
| | - Satoshi Tamaru
- Clinical Research Support Center, Mie University Hospital, Tsu 514-8507, Mie, Japan;
| | - Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan; (E.W.); (T.S.); (S.O.); (J.K.)
- Mie University Research Center for Cilia and Diseases, Tsu 514-8507, Mie, Japan
| |
Collapse
|
13
|
Kunitatsu K, Yamamoto Y, Nasu S, Taniji A, Kawashima S, Yamagishi N, Ito T, Inoue S, Kanai Y. Novel Peritoneal Sclerosis Rat Model Developed by Administration of Bleomycin and Lansoprazole. Int J Mol Sci 2023; 24:16108. [PMID: 38003303 PMCID: PMC10671295 DOI: 10.3390/ijms242216108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
In our preliminary experiment, peritoneal sclerosis likely induced by peritoneal dialysis was unexpectedly observed in the livers of rats given bleomycin and lansoprazole. We examined whether this peritoneal thickening around the liver was time-dependently induced by administration of both drugs. Male Wistar rats were injected with bleomycin and/or lansoprazole for 2 or 4 weeks. The 3YB-1 cell line derived from rat fibroblasts was treated by bleomycin and/or lansoprazole for 24 h. The administration of both drugs together, but not individually, thickened the peritoneal tissue around the liver. There was accumulation of collagen fibers, macrophages, and eosinophils under mesothelial cells. Expressions of Col1a1, Mcp1 and Mcp3 genes were increased in the peritoneal tissue around the liver and in 3YB-1 cells by the administration of both drugs together, and Opn genes had increased expressions in this tissue and 3YB-1 cells. Mesothelial cells indicated immunoreactivity against both cytokeratin, a mesothelial cell marker, and αSMA, a fibroblast marker, around the livers of rats given both drugs. Administration of both drugs induced the migration of macrophages and eosinophils and induced fibrosis associated with the possible activation of fibroblasts and the possible promotion of the mesothelial-mesenchymal transition. This might become a novel model of peritoneal sclerosis for peritoneal dialysis.
Collapse
Affiliation(s)
- Kosei Kunitatsu
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Yuta Yamamoto
- Department of Anatomy and Cell Biology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Shota Nasu
- Department of Anatomy and Cell Biology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Akira Taniji
- Department of Anatomy and Cell Biology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Shuji Kawashima
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Naoko Yamagishi
- Department of Anatomy and Cell Biology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Takao Ito
- Department of Anatomy and Cell Biology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Shigeaki Inoue
- Department of Emergency and Critical Care Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Yoshimitsu Kanai
- Department of Anatomy and Cell Biology, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| |
Collapse
|
14
|
Hassanein EHM, Kamel EO, Gad-Elrab WM, Ahmed MA, Mohammedsaleh ZM, Ali FEM. Lansoprazole attenuates cyclophosphamide-induced cardiopulmonary injury by modulating redox-sensitive pathways and inflammation. Mol Cell Biochem 2023; 478:2319-2335. [PMID: 36717473 PMCID: PMC10520119 DOI: 10.1007/s11010-023-04662-x] [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: 02/26/2022] [Accepted: 01/09/2023] [Indexed: 02/01/2023]
Abstract
Cyclophosphamide (CPA) is a classical chemotherapeutic drug widely used as an anticancer and immunosuppressive agent. However, it is frequently associated with significant toxicities to the normal cells of different organs, including the lung and heart. Lansoprazole (LPZ), a proton pump inhibitor (PPI), possesses antioxidant and anti-inflammatory properties. The current study investigated how LPZ protects against CPA-induced cardiac and pulmonary damage, focusing on PPARγ, Nrf2, HO-1, cytoglobin, PI3K/AKT, and NF-κB signaling. Animals were randomly assigned into four groups: normal control group (received vehicle), LPZ only group (Rats received LPZ at a dose of 50 mg/kg/day P.O. for 10 days), CPA group (CPA was administered (200 mg/kg) as a single i.p. injection on the 7th day), and cotreatment group (LPZ plus CPA). Histopathological and biochemical analyses were conducted. Our results revealed that LPZ treatment revoked CPA-induced heart and lung histopathological alterations. Also, LPZ potently mitigated CPA-induced cardiac and pulmonary oxidative stress through the activation of PPARγ, Nrf2/HO-1, cytoglobin, and PI3K/AKT signaling pathways. Also, LPZ effectively suppressed inflammatory response as evidenced by down-regulating the inflammatory strategic controller NF-κB, MPO, and pro-inflammatory cytokines. The present findings could provide a mechanistic basis for understanding LPZ's role in CPA-induced cardiopulmonary injury through the alleviation of oxidative stress and inflammatory burden.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Esam O Kamel
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Al-Azhar University, Assiut, Egypt
| | - Wail M Gad-Elrab
- Department of Human Anatomy & Embryology, Faculty of Medicine, Al-Azhar University, Assiut, Egypt
| | - Mohammed A Ahmed
- Pathology Department, Faculty of Medicine, Al-Azhar University, Assiut, Egypt
| | - Zuhair M Mohammedsaleh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, 71491, Kingdom of Saudi Arabia
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| |
Collapse
|
15
|
Ali FEM, Hassanein EHM, El-Bahrawy AH, Hemeda MS, Atwa AM. Neuroprotective effect of lansoprazole against cisplatin-induced brain toxicity: Role of Nrf2/ARE and Akt/P53 signaling pathways. J Chem Neuroanat 2023; 132:102299. [PMID: 37271475 DOI: 10.1016/j.jchemneu.2023.102299] [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: 02/17/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
Cisplatin is a chemotherapeutic agent usually used in treating different patterns of malignancies. One of the significant apparent complications of cisplatin chemotherapy is brain toxicity. The present study was conducted to evaluate the protective effects of lansoprazole on cisplatin-induced cortical intoxication. Thirty-two rats were allocated into four groups (8 rats/group); group I: received only a vehicle for 10 days, group II: lansoprazole was administered (50 mg/kg) via oral gavage for 10 days, group III: On 5th day of the experiment, rats were given cisplatin (10 mg/kg) i.p. once to induce cortical injury. Group IV: rats were given lansoprazole for 5 days before cisplatin and 5 days afterward. Lansoprazole administration significantly improved cisplatin-induced behavioral changes, as evidenced by decreasing the immobility time in forced swimming and open field tests. Besides, lansoprazole improved cortical histological changes, restored cortical redox balance, enhanced Nrf2/ARE expression, cisplatin-induced neuronal apoptosis, and dampened cisplatin inflammation. In addition, lansoprazole modulated cortical Akt/p53 signal. The present work was the first to show that lansoprazole co-administration reduced cortical toxicity in cisplatin-treated rats via multiple signaling pathways. The current findings provided crucial information for developing novel protective strategies to reduce cisplatin cortical toxicity.
Collapse
Affiliation(s)
- Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Ali H El-Bahrawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Mohamed S Hemeda
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| |
Collapse
|
16
|
Habotta OA, Abdeen A, El-Hanafy AA, Yassin N, Elgameel D, Ibrahim SF, Abdelrahaman D, Hasan T, Imbrea F, Ghamry HI, Fericean L, Behairy A, Atwa AM, Abdelkader A, Mahdi MR, El-Mosallamy SA. Sesquiterpene nootkatone counteracted the melamine-induced neurotoxicity via repressing of oxidative stress, inflammatory, and apoptotic trajectories. Biomed Pharmacother 2023; 165:115133. [PMID: 37454594 DOI: 10.1016/j.biopha.2023.115133] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
Melamine (ML), a chemical substance of high nitrogen content, is used as a food adulterant. Former evidences implied that ML could induce a variety of toxic effects including neurotoxicity and cognitive impairment. Therefore, the aim of this study was to delineate the protective effect of the nootkatone (NK) against ML-induced neural adverse effects. Rats were orally pretreated with NK (5 and 10 mg/kg) prior to the oral administration of ML (700 mg/kg) for a period of 28 days. Our findings unveiled remarkable alleviating effect of NK on MK-induced neurobehavioral disturbance in open field test. Furthermore, NK lessened ML-caused increases in the acetylcholine esterase level in the brain tissue of exposed rats. NK also decreased the neural oxidative stress as represented by elevated levels of SOD, CAT, and GSH along with decreased MDA and NO levels. Upregulated mRNA expression levels of neural NRF-2 and HO-1 were noticed after NK administration. Remarkable anti-inflammatory impact was prominent by decreased neural IL-1β, and TNF-α along with downregulated NF-κB and TLR-4 gene expression levels in NK-treated rats. Noteworthily, pre-treatment with NK decreased the immune reaction of RAGE and HMGB-1 induced by oral ML exposure. Brain histological examination validated the obtained biochemical and molecular results. To sum up, these outcomes reveal that NK successfully alleviated the neural damage induced by ML via blocking of oxidative stress, and inflammatory signaling pathways. Consequently, our study may suggest NK as a new effective therapeutic supplement for treatment of ML-mediated neurotoxicity in rats via inhibition of HMGB-1-RAGE/TLR-4/NF-κB.
Collapse
Affiliation(s)
- Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Ahmed Abdeen
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt.
| | - Aya A El-Hanafy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt; Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, New Mansoura University, New Mansoura, 35516, Egypt.
| | - Neimet Yassin
- Department of Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Dina Elgameel
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Tanta University, Tanta 31111, Egypt.
| | - Samah F Ibrahim
- Department of Clinical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Doaa Abdelrahaman
- Department of Clinical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Tabinda Hasan
- Department of Basic Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Florin Imbrea
- Department of Crop Science, Faculty of Agriculture, University of Life Sciences "King Mihai I" from Timisoara, 119, Calea Aradului, 300645 Timisoara, Romania.
| | - Heba I Ghamry
- Nutrition and Food Sciences, Department of Home Economics, Faculty of Home Economics, King Khalid University, P.O. Box 960, Abha 61421, Saudi Arabia.
| | - Liana Fericean
- Department of Biology and Plant protection, Faculty of Agriculture. University of Life Sciences "King Michael I" from Timișoara, Calea Aradului 119, CUI 3487181, Romania.
| | - Ali Behairy
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha 13518, Egypt.
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo 11829, Egypt.
| | - Afaf Abdelkader
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha 13518, Egypt.
| | - Mohamed R Mahdi
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt; Department of Basic Medical Sciences, Faculty of Medicine, Galala University, Suez, Egypt.
| | - Shaaban A El-Mosallamy
- Department of Forensic Medicine and Clinical toxicology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
17
|
Alqahtani MJ, Negm WA, Saad HM, Salem EA, Hussein IA, Ibrahim HA. Fenofibrate and Diosmetin in a rat model of testicular toxicity: New insight on their protective mechanism through PPAR-α/NRF-2/HO-1 signaling pathway. Biomed Pharmacother 2023; 165:115095. [PMID: 37413905 DOI: 10.1016/j.biopha.2023.115095] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023] Open
Abstract
One of the most significant chemotherapeutic side effects of cisplatin (Cis) that limits its use and efficacy is testicular toxicity. Thus, the objective of the present study was to investigate the possible ameliorative effect of Fenofibrate (Fen), Diosmetin (D), and their combination against cis-mediated testicular damage. Fifty-four adult male albino rats were randomly allocated into nine groups (6 rats each): Control group, Fen (100 mg/kg), D20 (20 mg/kg), D40 (40 mg/kg), Cis group (7 mg/kg), Cis +Fen group (7 mg/kg+100 mg/kg), Cis+D20 group (7 mg/kg+20 mg/kg), Cis+D40 group (7 mg/kg+40 mg/kg), Cis+Fen+D40 treated group (7 mg/kg+100 mg/kg+40 mg/kg). Relative testicular weight, epididymal sperm count and viability, serum testosterone level, testicular oxidative stress indices, mRNA expression of peroxisome proliferator-activated receptor alpha (PPAR-α), nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1), histopathological, and immunohistochemical alterations were assessed. Our results revealed that cis administration induced testicular oxidative and inflammatory damage as indicated by a substantial reduction in relative testicular weight, sperm parameters, serum testosterone levels, the antioxidant enzyme activity of catalase, and Johnson's histopathological score, PPAR-α/NRF-2/HO-1 and proliferating cell nuclear antigen (PCNA) immunoexpression with marked increment in malondialdehyde (MDA), Cosentino's score, nuclear factor kappa B (NF-κβ p65), interleukin (IL)- 1β and caspase 3 in testicular tissue. Interestingly, Fen and D diminished the harmful effects of cis on testes via upregulation of the antioxidant activities and downregulation of lipid peroxidation, apoptosis, and inflammation. Moreover, the combination therapy Fen/D40 also exhibited a more pronounced enhancement of previous markers than either treatment alone. In conclusion, because of their antioxidant, anti-inflammatory, and anti-apoptotic properties, cotreatment with Fen or D or their combination could be beneficial in reducing the harmful impacts of cis on testicular tissue, particularly in patients that receive cis chemotherapy.
Collapse
Affiliation(s)
- Moneerah J Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia.
| | - Walaa A Negm
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of veterinary medicine, Matrouh University, Marsa Matrouh, Egypt.
| | - Esraa A Salem
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Shebeen ElKom 32511, Egypt.
| | - Ismail A Hussein
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt.
| | - Hanaa A Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Tanta, Egypt.
| |
Collapse
|
18
|
Yamagishi N, Yamamoto Y, Nishi T, Ito T, Kanai Y. Lansoprazole protects hepatic cells against cisplatin-induced oxidative stress through the p38 MAPK/ARE/Nrf2 pathway. PLoS One 2023; 18:e0287788. [PMID: 37384717 PMCID: PMC10309994 DOI: 10.1371/journal.pone.0287788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023] Open
Abstract
Lansoprazole, a proton pump inhibitor, can exert antioxidant effects through the induction of the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, independently of the inhibition of acid secretion in the gastrointestinal tract. Lansoprazole has been reported to provide hepatoprotection in a drug-induced hepatitis animal model through the Nrf2/heme oxygenase-1 (HO1) pathway. We sought to investigate the molecular mechanism of cytoprotection by lansoprazole. An in vitro experimental model was conducted using cultured rat hepatic cells treated with lansoprazole to analyze the expression levels of Nrf2 and its downstream genes, the activity of Nrf2 using luciferase reporter assays, cisplatin-induced cytotoxicity, and signaling pathways involved in Nrf2 activation. Lansoprazole treatment of rat liver epithelial RL34 cells induced transactivation of Nrf2 and the expression of the Nrf2-dependent antioxidant genes encoding HO1, NAD(P)H quinone oxidoreductase-1, and glutathione S-transferase A2. Furthermore, cycloheximide chase experiments revealed that lansoprazole prolongs the half-life of the Nrf2 protein. Notably, cell viability was significantly increased by lansoprazole treatment in a cisplatin-induced cytotoxicity model. Moreover, the siRNA knockdown of Nrf2 fully abolished the cytoprotective effect of lansoprazole, whereas the inhibition of HO1 by tin-mesoporphyrin only partially abolished this. Finally, lansoprazole promoted the phosphorylation of p38 mitogen-activated protein kinase (MAPK) but not that of the extracellular signal-regulated kinase or the c-Jun N-terminal kinase. Using SB203580, a specific inhibitor for p38 MAPK, the lansoprazole-induced Nrf2/antioxidant response elements pathway activation and cytoprotective effects were shown to be exclusively p38 MAPK dependent. Lansoprazole was shown by these results to exert a cytoprotective effect on liver epithelial cells against the cisplatin-induced cytotoxicity through the p38 MAPK signaling pathway. This could have potential applications for the prevention and treatment of oxidative injury in the liver.
Collapse
Affiliation(s)
- Naoko Yamagishi
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuta Yamamoto
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Toshio Nishi
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Takao Ito
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yoshimitsu Kanai
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
19
|
El-Beheiry KM, El-Sayed El-Sayad M, El-Masry TA, Elsisi AE. Combination of metformin and hesperidin mitigates cyclophosphamide-induced hepatotoxicity. Emerging role of PPAR-γ/Nrf-2/NF-κB signaling pathway. Int Immunopharmacol 2023; 117:109891. [PMID: 36812672 DOI: 10.1016/j.intimp.2023.109891] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/02/2023] [Accepted: 02/11/2023] [Indexed: 02/22/2023]
Abstract
Cyclophosphamide (CP) is widely used as an immunosuppressive and chemotherapeutic drug. However, its therapeutic application is restricted by its adverse effects, particularly hepatotoxicity. Both metformin (MET) and hesperidin (HES) have promising antioxidant, anti-inflammatory, and anti-apoptotic effects. Therefore, the principal aim of the current study is to investigate the hepatoprotective effects of MET, HES, and their combinations on the CP-induced hepatotoxicity model. Hepatotoxicity was evoked by a single (I.P) injection of CP (200 mg/kg) on day 7. For this study, 64 albino rats were randomly categorized into eight equal groups; naïve, control vehicle, untreated CP (200 mg/kg, IP), and CP 200 groups treated with MET 200, HES 50, HES 100 or a combination of MET 200 with HES 50 and HES 100 respectively orally daily for 12 days. At the end of the study, the liver function biomarkers, oxidative stress, inflammatory parameters, histopathological and immunohistochemical analysis of PPAR-γ, Nrf-2, NF-κB, Bcl-2, and caspase3 were assessed. CP significantly increased serum ALT, AST, total bilirubin, hepatic MDA, NO content, NF-κB, and TNF-α. Otherwise, albumin, hepatic GSH content, Nrf-2, and PPAR-γ expression decreased considerably compared to the control vehicle group. The combinations of MET200 with HES50 or HES100 induced pronounced hepatoprotective, anti-oxidative, anti-inflammatory, and anti-apoptotic effects on CP-treated rats. The possible explanation of such hepatoprotective effects may be mediated via upregulation of Nrf-2, PPAR-γ, Bcl-2 expression, hepatic GSH content, and marked suppression of TNF-α and NF-κB expression. In conclusion, the current study showed that combining MET and HES revealed a remarkable hepatoprotective effect against CP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Kareman M El-Beheiry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Magda El-Sayed El-Sayad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Alaa E Elsisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
20
|
Dai C, Li M, Liu Y, Tran DH, Jiang H, Tang S, Shen J. Involvement of the inhibition of mitochondrial apoptotic, p53, NF-κB pathways and the activation of Nrf2/HO-1 pathway in the protective effects of curcumin against copper sulfate-induced nephrotoxicity in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114480. [PMID: 38321692 DOI: 10.1016/j.ecoenv.2022.114480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/29/2022] [Accepted: 12/23/2022] [Indexed: 02/08/2024]
Abstract
Chronic copper exposure could cause potential nephrotoxicity and effective therapy strategies are limited. This study investigated the protective effects of curcumin on copper sulfate (CuSO4)-induced renal damage in a mouse model and the underlying molecular mechanisms. Mice were administrated orally with CuSO4 (100 mg/kg per day) in combination with or without curcumin (50, 100 or 200 mg/kg per day, orally) for 28 days. Results showed that curcumin supplementation significantly reduce the Cu accumulation in the kidney tissues of mice and improved CuSO4-induced renal dysfunction. Furthermore, curcumin supplantation also significantly ameliorated Cu exposure-induced oxidative stress and tubular necrosis in the kidneys of mice. Moreover, compared to the CuSO4 alone group, curcumin supplementation at 200 mg/kg per day significantly decreased CuSO4-induced the expression of p53, Bax, IL-1β, IL-6, and TNF-α proteins, levels of NF-κB mRNA, levels of caspases-9 and - 3 activities, and cell apoptosis, and significantly increased the levels of Nrf2 and HO-1 mRNAs in the kidney tissues. In conclusion, for the first time, our results reveal that curcumin could trigger the inhibition of oxidative stress, mitochondrial apoptotic, p53, and NF-κB pathways and the activation of Nrf2/HO-1 pathway to ameliorate Cu overload-induced nephrotoxicity in a mouse model. Our study highlights that curcumin supplementation may be a promising treatment strategy for treating copper overload-caused nephrotoxicity.
Collapse
Affiliation(s)
- Chongshan Dai
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, PR China; Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing 100193, PR China.
| | - Meng Li
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Yue Liu
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Diem Hong Tran
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Haiyang Jiang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, PR China; Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing 100193, PR China
| | - Shusheng Tang
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Jianzhong Shen
- College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, PR China; Beijing Key Laboratory of Detection Technology for Animal-Derived Food Safety, Beijing 100193, PR China
| |
Collapse
|
21
|
Ala M. Sestrin2 Signaling Pathway Regulates Podocyte Biology and Protects against Diabetic Nephropathy. J Diabetes Res 2023; 2023:8776878. [PMID: 36818747 PMCID: PMC9937769 DOI: 10.1155/2023/8776878] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/22/2022] [Accepted: 02/04/2023] [Indexed: 02/12/2023] Open
Abstract
Sestrin2 regulates cell homeostasis and is an upstream signaling molecule for several signaling pathways. Sestrin2 leads to AMP-activated protein kinase- (AMPK-) and GTPase-activating protein activity toward Rags (GATOR) 1-mediated inhibition of mammalian target of rapamycin complex 1 (mTORC1), thereby enhancing autophagy. Sestrin2 also improves mitochondrial biogenesis via AMPK/Sirt1/peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1α) signaling pathway. Blockade of ribosomal protein synthesis and augmentation of autophagy by Sestrin2 can prevent misfolded protein accumulation and attenuate endoplasmic reticulum (ER) stress. In addition, Sestrin2 enhances P62-mediated autophagic degradation of Keap1 to release nuclear factor erythroid 2-related factor 2 (Nrf2). Nrf2 release by Sestrin2 vigorously potentiates antioxidant defense in diabetic nephropathy. Impaired autophagy and mitochondrial biogenesis, severe oxidative stress, and ER stress are all deeply involved in the development and progression of diabetic nephropathy. It has been shown that Sestrin2 expression is lower in the kidney of animals and patients with diabetic nephropathy. Sestrin2 knockdown aggravated diabetic nephropathy in animal models. In contrast, upregulation of Sestrin2 enhanced autophagy, mitophagy, and mitochondrial biogenesis and suppressed oxidative stress, ER stress, and apoptosis in diabetic nephropathy. Consistently, overexpression of Sestrin2 ameliorated podocyte injury, mesangial proliferation, proteinuria, and renal fibrosis in animal models of diabetic nephropathy. By suppressing transforming growth factor beta (TGF-β)/Smad and Yes-associated protein (YAP)/transcription enhancer factor 1 (TEF1) signaling pathways in experimental models, Sestrin2 hindered epithelial-mesenchymal transition and extracellular matrix accumulation in diabetic kidneys. Moreover, modulation of the downstream molecules of Sestrin2, for instance, augmentation of AMPK or Nrf2 signaling and inhibition of mTORC1, has been protective in diabetic nephropathy. Regarding the beneficial effects of Sestrin2 on diabetic nephropathy and its interaction with several signaling molecules, it is worth targeting Sestrin2 in diabetic nephropathy.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
22
|
Hashemi M, Azarnia M, Hajebrahimi Z, Nejad Ebrahimi S. The effect of hydroalcoholic extract of Cichorium intybus leaf on aryl hydrocarbon receptor expression in the testis of Wistar rats exposed to cigarette smoke. AVICENNA JOURNAL OF PHYTOMEDICINE 2023; 13:58-69. [PMID: 36698732 PMCID: PMC9840776 DOI: 10.22038/ajp.2022.21307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/27/2022] [Accepted: 09/23/2022] [Indexed: 01/27/2023]
Abstract
Objective Cigarette smoke (CS) contains compounds such as reactive oxygen species (ROS). Oxidative stress caused by excessive ROS eventually leads to germ cell apoptosis and male infertility. The leaves of Cichorium intybus (chicory) are rich in natural antioxidants, but their protective effects on the adverse effects of CS on testicular tissue have not been studied. Materials and Methods 24 Wistar rats were classified into four groups: control, extract: treatment with chicory extract (200 mg/kg body weight/day) for 13 weeks, smoke: exposed to CS for 13 weeks, and smoke + extract: exposed to CS and treated with the C. intybus extract. Histological and biochemical analyses and apoptosis assay were done, and Ahr, and Cyp1a1 expression was determined. Results Treatment with C. intybus compensated for the reduction of Sertoli cells, spermatogonia, spermatocytes, and spermatids caused by CS. Chicory extract reduced free radicals and improved antioxidant status. The lowest and highest percentage of apoptotic cells was observed in the extract and smoke groups, respectively, while simultaneous treatment with C. intybus extract led to a significant reduction of apoptotic cells. The mean Ahr levels in the control, extract, smoke and smoke + extract groups were 1.00±0.57, 1.93±0.25, 5.98±0.42, and 0.62±0.22, respectively (p˂0.05). The mean levels of Cyp1a1 expression in the control, extract, smoke and smoke + extract groups were 1.00±0.31, 2.28±0.65, 5.55±0.40, and 0.21±0.23 (p˂0.05). Conclusion The C. intybus extract probably affected Cyp1a1 expression by downregulation of Ahr. These led to a decrease in free radicals and apoptosis, and an improvement in antioxidant status.
Collapse
Affiliation(s)
- Maryam Hashemi
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Mahnaz Azarnia
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Zahra Hajebrahimi
- A&S Research Institute, Ministry of Science Research and Technology, Tehran, IranDepartment of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G. C., Evin, Tehran, IranDepartment of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G. C., Evin, Tehran, Iran Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G. C., Evin, Tehran, Iran Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G. C., Evin, Tehran, Iran
| | - Samad Nejad Ebrahimi
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, G. C., Evin, Tehran, Iran
| |
Collapse
|
23
|
Dai C, Tian E, Hao Z, Tang S, Wang Z, Sharma G, Jiang H, Shen J. Aflatoxin B1 Toxicity and Protective Effects of Curcumin: Molecular Mechanisms and Clinical Implications. Antioxidants (Basel) 2022; 11:antiox11102031. [PMID: 36290754 PMCID: PMC9598162 DOI: 10.3390/antiox11102031] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/09/2022] [Accepted: 10/10/2022] [Indexed: 11/26/2022] Open
Abstract
One of the most significant classes of mycotoxins, aflatoxins (AFTs), can cause a variety of detrimental outcomes, including cancer, hepatitis, aberrant mutations, and reproductive issues. Among the 21 identified AFTs, aflatoxin B1 (AFB1) is the most harmful to humans and animals. The mechanisms of AFB1-induced toxicity are connected to the generation of excess reactive oxygen species (ROS), upregulation of CYP450 activities, oxidative stress, lipid peroxidation, apoptosis, mitochondrial dysfunction, autophagy, necrosis, and inflammatory response. Several signaling pathways, including p53, PI3K/Akt/mTOR, Nrf2/ARE, NF-κB, NLRP3, MAPKs, and Wnt/β-catenin have been shown to contribute to AFB1-mediated toxic effects in mammalian cells. Curcumin, a natural product with multiple therapeutic activities (e.g., anti-inflammatory, antioxidant, anticancer, and immunoregulation activities), could revise AFB1-induced harmful effects by targeting these pathways. Therefore, the potential therapeutic use of curcumin against AFB1-related side effects and the underlying molecular mechanisms are summarized. This review, in our opinion, advances significant knowledge, sparks larger discussions, and drives additional improvements in the hazardous examination of AFTs and detoxifying the application of curcumin.
Collapse
Affiliation(s)
- Chongshan Dai
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
- Correspondence:
| | - Erjie Tian
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471023, China
| | - Zhihui Hao
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Shusheng Tang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Zhanhui Wang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Gaurav Sharma
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Haiyang Jiang
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jianzhong Shen
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
24
|
Elsisi AE, Elmarhoumy EH, Osman EY. Protective effect of cilostazol and verapamil against thioacetamide-induced hepatotoxicity in rats may involve Nrf2/GSK-3β/NF-κB signaling pathway. Toxicol Res (Camb) 2022; 11:718-729. [PMID: 36337252 PMCID: PMC9618097 DOI: 10.1093/toxres/tfac045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Verapamil (VER) and cilostazol (Cilo) are mostly used as cardiovascular drugs; they have beneficial effects on different organs toxicities. AIM we investigated whether the Nuclear factor erythroid 2-related factor 2 (Nrf2), Glycogen synthase kinase-3β (GSK-3β), and Nuclear factor-kappa B (NF-κB) pathway involved in the protective role of these drugs against Thioacetamide (TAA) induced hepatotoxicity. METHOD male rats were randomized divided into five groups, each group (n = 10): control, TAA, VER+TAA, Cilo+TAA, and VER+Cilo+TAA groups. Hepatotoxicity induced in rats by TAA injection once on the 7th day of the experiment. RESULTS TAA-induced hepatotoxicity indicated by a significant elevated in serum markers (Alanine aminotransferases (ALT), Aspartate aminotransferases (AST), and bilirubin), oxidative stress markers (Malondialdehyde (MDA), and Nitric oxide (NO)), and protein levels markers (NF-κB, and S100 calcium-binding protein A4 (S100A4)). Also, TAA decreased Nrf2, and increased GSK-3β genes expression. Histopathological alterations in the liver also appeared as a response to TAA injection. On the other hand VER and/or Cilo significantly prevented TAA-induced hepatotoxicity in rats through significantly decreased in ALT, AST, bilirubin, MDA, NO, NF-κB, and S100A4 protein levels. Also, they increased Nrf2 and decreased GSK-3β genes expression which caused improvement in the histopathological changes of the liver. CONCLUSION the addition of verapamil to cilostazol potentiated the hepatoprotective activity, and inhibited the progression of hepatotoxicity caused by TAA through the Nrf2/GSK-3β/NF-κBpathway and their activity on oxidative stress, inflammation, and NF-κB protein expression.
Collapse
Affiliation(s)
- Alaa E Elsisi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Esraa H Elmarhoumy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Enass Y Osman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
25
|
Assar DH, Asa SA, El-Abasy MA, Elbialy ZI, Shukry M, Latif AAE, BinMowyna MN, Althobaiti NA, El-Magd MA. Aspergillus awamori attenuates ochratoxin A-induced renal and cardiac injuries in rabbits by activating the Nrf2/HO-1 signaling pathway and downregulating IL1β, TNFα, and iNOS gene expressions. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:69798-69817. [PMID: 35576029 PMCID: PMC9512883 DOI: 10.1007/s11356-022-20599-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/29/2022] [Indexed: 05/10/2023]
Abstract
Ochratoxin A (OTA) is one of the most dangerous and that pollute agricultural products, inducing a variety of toxic effects in humans and animals. The current study explored the protective effect of different concentrations of Aspergillus awamori (A. awamori) against OTA (0.3 mg/kg diet) induced renal and cardiac damage by exploring its mechanism of action in 60 New Zealand white male rabbits. Dietary supplementation of A. awamori at the selected doses of 50, 100, and 150 mg/kg diet, respectively, for 2 months significantly improved the rabbit's growth performance; modulated the suppressed immune response and restored the altered hematological parameters; reduced the elevated levels of renal injury biomarkers such as urea, creatinine, and alkaline phosphatase; and increased serum total proteins concentrations. Moreover, it also declined enzymatic activities of cardiac injury biomarkers, including AST, LDH, and CK-MB. A. awamori alleviated OTA-induced degenerative and necrotic changes in the kidney and heart of rabbits. Interestingly, A. awamori upregulated Nrf2/OH-1 signaling pathway. Therefore enhanced TAC, CAT, and SOD enzyme activities and reduced OTA-induced oxidative and nitrosative stress by declining iNOS gene expression and consequently lowered MDA and NO levels. In addition to attenuating renal and cardiac inflammation via reducing IL-1β, TNF-α gene expressions in a dose-dependent response. In conclusion,this is the first report to pinpoint that dietary incorporation of A. awamori counteracted OTA-induced renal and cardiac damage by potentiating the rabbit's antioxidant defense system through its potent antioxidant, free radical scavenging, and anti-inflammatory properties in a dose-dependent response. Based on our observations, A. awamori could be utilized as a natural protective agent against ochratoxicosis in rabbits.
Collapse
Affiliation(s)
- Doaa H. Assar
- Clinical Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Samah Abou Asa
- Pathology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Moshira A. El-Abasy
- Poultry and Rabbit Diseases Department, Faculty of Veterinary Medicine, Kafr El-Sheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Zizy I. Elbialy
- Fish Processing and Biotechnology Department, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, 33516 Kafr El-Sheikh, Egypt
| | - Mustafa Shukry
- Physiology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Amera Abd El Latif
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| | - Mona N. BinMowyna
- College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Norah A. Althobaiti
- Biology Department, College of Science and Humanities-Al Quwaiiyah, Shaqra University, Al Quwaiiyah, 19257 Saudi Arabia
| | - Mohammed A. El-Magd
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Sheikh, 33516 Egypt
| |
Collapse
|
26
|
Amano Y, Yamane M, Honda H. RAID: Regression Analysis–Based Inductive DNA Microarray for Precise Read-Across. Front Pharmacol 2022; 13:879907. [PMID: 35935858 PMCID: PMC9354856 DOI: 10.3389/fphar.2022.879907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/30/2022] [Indexed: 12/02/2022] Open
Abstract
Chemical structure-based read-across represents a promising method for chemical toxicity evaluation without the need for animal testing; however, a chemical structure is not necessarily related to toxicity. Therefore, in vitro studies were often used for read-across reliability refinement; however, their external validity has been hindered by the gap between in vitro and in vivo conditions. Thus, we developed a virtual DNA microarray, regression analysis–based inductive DNA microarray (RAID), which quantitatively predicts in vivo gene expression profiles based on the chemical structure and/or in vitro transcriptome data. For each gene, elastic-net models were constructed using chemical descriptors and in vitro transcriptome data to predict in vivo data from in vitro data (in vitro to in vivo extrapolation; IVIVE). In feature selection, useful genes for assessing the quantitative structure–activity relationship (QSAR) and IVIVE were identified. Predicted transcriptome data derived from the RAID system reflected the in vivo gene expression profiles of characteristic hepatotoxic substances. Moreover, gene ontology and pathway analysis indicated that nuclear receptor-mediated xenobiotic response and metabolic activation are related to these gene expressions. The identified IVIVE-related genes were associated with fatty acid, xenobiotic, and drug metabolisms, indicating that in vitro studies were effective in evaluating these key events. Furthermore, validation studies revealed that chemical substances associated with these key events could be detected as hepatotoxic biosimilar substances. These results indicated that the RAID system could represent an alternative screening test for a repeated-dose toxicity test and toxicogenomics analyses. Our technology provides a critical solution for IVIVE-based read-across by considering the mode of action and chemical structures.
Collapse
|
27
|
Alipour M, Hajipour-Verdom B, Javan M, Abdolmaleki P. Static and Electromagnetic Fields Differently Affect Proliferation and Cell Death Through Acid Enhancement of ROS Generation in Mesenchymal Stem Cells. Radiat Res 2022; 198:384-395. [PMID: 35867630 DOI: 10.1667/rade-21-00037.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/17/2022] [Indexed: 11/03/2022]
Abstract
Magnetic fields remotely influence cellular homeostasis as a physical agent through the changes in cell physicochemical reactions. Magnetic fields affect cell fate, which may provide an important and interesting challenge in stem cell behaviors. Here, we investigated the effects of the static magnetic field (SMF, 20 mT) and electromagnetic field (EMF, 20 mT-50 Hz) on reactive oxygen species (ROS) production and the acidic pH conditions as stimuli to change cell cycle progression and cell death in mesenchymal stem cells. Results show that SMF, EMF, and their simultaneous (SMF+EMF) administration increase ROS and expression of nuclear factor erythroid 2-related factor 2 (Nrf2), superoxide dismutase 2 (SOD2), and glutathione-S-transferase (GST) as an antioxidant defense system. Besides, intracellular pH (pHi) decreases in presence of either EMF or SMF+EMF, but not SMF. Decreased ROS content using ascorbic acid in these treatments leads to increased pH compared to the magnetic field treatments alone. Furthermore, each magnetic field has different effects on the cellular process of stem cells, including cell cycle, apoptosis and necrosis. Moreover, treatment by SMF enhances the cell viability after 24 h, while EMF or SMF+EMF decreases it. These observations indicate that fluctuations of ROS generation and acid enhancement during SMF and EMF treatments may reveal their beneficial and adverse effects on the molecular and cellular mechanisms involved in the growth, death, and differentiation of stem cells.
Collapse
Affiliation(s)
| | | | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, 14115-154, Iran
| | | |
Collapse
|
28
|
Castro MP. Unintended effects of proton pump inhibitors (PPIs) in patients with glioblastoma (GBM): A double-edged sword. Neurooncol Pract 2022; 9:344-345. [PMID: 35855453 PMCID: PMC9290858 DOI: 10.1093/nop/npac035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Michael P Castro
- Beverly Hills Cancer Center , Beverly Hills, California , USA
- Personalized Cancer Medicine, PLLC Los Angeles, California , USA
- Cellworks Group, Inc. S. San Francisco, California , USA
| |
Collapse
|
29
|
Gupta V, Hammond CL, Roztocil E, Gonzalez MO, Feldon SE, Woeller CF. Thinking inside the box: Current insights into targeting orbital tissue remodeling and inflammation in thyroid eye disease. Surv Ophthalmol 2022; 67:858-874. [PMID: 34487739 PMCID: PMC8891393 DOI: 10.1016/j.survophthal.2021.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 12/21/2022]
Abstract
Thyroid eye disease (TED) is an autoimmune disorder that manifests in the orbit. In TED, the connective tissue behind the eye becomes inflamed and remodels with increased fat accumulation and/or increased muscle and scar tissue. As orbital tissue expands, patients develop edema, exophthalmos, diplopia, and optic neuropathy. In severe cases vision loss may occur secondary to corneal scarring from exposure or optic nerve compression. Currently there is no cure for TED, and treatments are limited. A major breakthrough in TED therapy occurred with the FDA approval of teprotumumab, a monoclonal insulin-like growth factor 1 receptor (IGF1R) blocking antibody. Yet, teprotumumab therapy has limitations, including cost, infusion method of drug delivery, variable response, and relapse. We describe approaches to target orbital fibroblasts and the complex pathophysiology that underlies tissue remodeling and inflammation driving TED. Further advances in the elucidation of the mechanisms of TED may lead to prophylaxis based upon early biomarkers as well as lead to more convenient, less expensive therapies.
Collapse
Affiliation(s)
- Vardaan Gupta
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Christine L Hammond
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Elisa Roztocil
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Mithra O Gonzalez
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Steven E Feldon
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA
| | - Collynn F Woeller
- Flaum Eye Institute, University of Rochester, 210 Crittenden Boulevard, Rochester, New York 14642, USA.
| |
Collapse
|
30
|
Hammond CL, Roztocil E, Gupta V, Feldon SE, Woeller CF. More than Meets the Eye: The Aryl Hydrocarbon Receptor is an Environmental Sensor, Physiological Regulator and a Therapeutic Target in Ocular Disease. FRONTIERS IN TOXICOLOGY 2022; 4:791082. [PMID: 35295218 PMCID: PMC8915869 DOI: 10.3389/ftox.2022.791082] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/08/2022] [Indexed: 12/22/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand activated transcription factor originally identified as an environmental sensor of xenobiotic chemicals. However, studies have revealed that the AHR regulates crucial aspects of cell growth and metabolism, development and the immune system. The importance of the AHR and AHR signaling in eye development, toxicology and disease is now being uncovered. The AHR is expressed in many ocular tissues including the retina, choroid, cornea and the orbit. A significant role for the AHR in age-related macular degeneration (AMD), autoimmune uveitis, and other ocular diseases has been identified. Ligands for the AHR are structurally diverse organic molecules from exogenous and endogenous sources. Natural AHR ligands include metabolites of tryptophan and byproducts of the microbiome. Xenobiotic AHR ligands include persistent environmental pollutants such as dioxins, benzo (a) pyrene [B (a) P] and polychlorinated biphenyls (PCBs). Pharmaceutical agents including the proton pump inhibitors, esomeprazole and lansoprazole, and the immunosuppressive drug, leflunomide, activate the AHR. In this review, we highlight the role of the AHR in the eye and discuss how AHR signaling is involved in responding to endogenous and environmental stimuli. We also present the emerging concept that the AHR is a promising therapeutic target for eye disease.
Collapse
Affiliation(s)
| | | | | | | | - Collynn F. Woeller
- Flaum Eye Institute, Rochester, NY, United States
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, United States
- *Correspondence: Collynn F. Woeller,
| |
Collapse
|
31
|
Aala J, Harchegani AB, Monsef HA, Mohsenifar Z, Ebrahimi P, Parvizi MR. N-Acetyl cysteine mitigates histopathological changes and inflammatory genes expressions in the liver of cadmium exposed rats. Environ Anal Health Toxicol 2021; 36:e2021024-0. [PMID: 34711021 PMCID: PMC8850162 DOI: 10.5620/eaht.2021024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 09/23/2021] [Indexed: 11/11/2022] Open
Abstract
This study aimed to consider the expression of Nrf2, NLRP3 and caspase 1 genes, as well as oxidative stress, and the protective role of N-acetyl cysteine (NAC) in the liver of rats treated with cadmium (Cd). Male rats were randomly divided into five groups including G1 (control), G2 (single dose of Cd), G3 (continuous dose of Cd), G4 (single dose of Cd + NAC), and G5 (continuous dose of Cd + NAC). Levels of malondialdehyde (MDA) and total antioxidant capacity (TAC) were measured. Expression of Nrf2, NLRP3 and caspase 1 genes was considered using RT-PCR. NAC treatments significantly improved TAC, but decreased MDA values in rats that exposed to continuous dose of Cd (p<0.05). Exposure to continuous dose of Cd caused a significant decrease in Nrf2 expression by 2.46-fold (p<0.001), but enhanced expression of NLRP3 and Caspase 1 genes by 3.13-fold and 3.16-fold), respectively (p<0.001). Compared to rats that treated to continuous dose of Cd, NAC supplementation enhanced the expression of Nrf2 by 1.67-fold (p<0.001) and reduced the expression of NLRP3 and Caspase 1 genes by 1.39-fold (p<0.001) and 1.58-fold (p<0.001), respectively. Down-regulation of Nrf2 and overexpression of NLRP3 and caspase 1 seems to be one of the main mechanisms of Cd toxicity on liver tissue. NAC protects liver tissue against Cd-induced oxidative injuries via enhancement of Nrf2 expression and reduction of NLRP3 and caspase 1 genes.
Collapse
Affiliation(s)
- Jalal Aala
- Department of Physiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Asghar Beigi Harchegani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hanieh Akhlaghi Monsef
- Department of Genetics, Faculty of New Sciences, Medical Science Branch, Islamic Azad University, Tehran, Iran
| | - Zhaleh Mohsenifar
- Ayatollah Taleghani Educational Hospital, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pirooz Ebrahimi
- University Scientific Education and Research Network, Tehran, Iran
| | - Mohammad Reza Parvizi
- Department of Physiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Cai Y, Li B, Peng D, Wang X, Li P, Huang M, Xing H, Chen J. Crm1-Dependent Nuclear Export of Bach1 is Involved in the Protective Effect of Hyperoside on Oxidative Damage in Hepatocytes and CCl 4-induced Acute Liver Injury. J Inflamm Res 2021; 14:551-565. [PMID: 33658828 PMCID: PMC7920627 DOI: 10.2147/jir.s279249] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
Background Nrf2-Bach1 antioxidant signaling pathway is considered as one of the most important mechanisms of cellular resistance to oxidative injury. The effect of hyperoside (Hyp) on the expression and distribution of Bach1, the relationship of Hyp's antioxidative effect and the influence of Bach1 remains unclear. Purpose The aim of this study was to investigate the role and mechanisms of Bach1 in the protective effect of Hyp on oxidative liver injury. Methods The protective effect of Hyp on oxidative stress injury was observed in vivo and in vitro. Next, the influence of Hyp on Bach1 expression and distribution, and competitive combination of Nrf2-Bach1 with ARE in H2O2-induced L02 cell was studied by Western blot, RT-PCR, immunofluorescence and CHIP assay. Finally, the expressions of Crm1, ERK and p38 and their roles on Hyp mediated nuclear export of Bach1 were investigated by Western blot. Results Hyp ameliorated the pathological damage, reduced the liver index, AST, ALT and MDA activities, and increased SOD and GSH levels in the CCl4-induced acute liver injury mouse model. Hyp attenuated H2O2-induced oxidative stress injury in L02 cells. Hyp promoted the early rapid redistribution of Bach1 from nucleus to cytoplasm. CHIP analyses demonstrated that Hyp enhanced the levels of Nrf2-ARE complex, and weakened the levels of Bach1-ARE complex within three hours. In addition, Hyp enhanced transport protein Crm1 expression and ERK1/2 activity. And LMB, a Crm1 inhibitor, attenuated the effect of Hyp on Bach1 nuclear export and anti-oxidation. U0126, an ERK1/2 inhibitor, reduced the effect of Hyp on Crm1 expression and the Bach1 redistribution. Conclusion The hepatoprotective mechanism of Hyp was related to improve Bach1 nuclear export depending on ERK1/2-Crm1 to upregulate the level of Nrf2 binding to ARE.
Collapse
Affiliation(s)
- Yongqing Cai
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Bin Li
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Dan Peng
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Xianfeng Wang
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Pan Li
- Department of Pharmacy, Fengdu Traditional Chinese Medicine Hospital, Chongqing, 408299, People's Republic of China
| | - Mingchun Huang
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, People's Republic of China
| | - Haiyan Xing
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Jianhong Chen
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| |
Collapse
|
33
|
Monir DM, Mahmoud ME, Ahmed OG, Rehan IF, Abdelrahman A. Forced exercise activates the NrF2 pathway in the striatum and ameliorates motor and behavioral manifestations of Parkinson's disease in rotenone-treated rats. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2020; 16:9. [PMID: 33158454 PMCID: PMC7646065 DOI: 10.1186/s12993-020-00171-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/16/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Parkinson's disease (PD) is a common neurodegenerative disorder characterized by progressive loss of nigrostriatal dopaminergic neurons leading to dopamine depletion and problems of movement, emotions, and cognition. While the pathogenesis of PD is not clear, damage of dopaminergic neurons by oxygen-derived free radicals is considered an important contributing mechanism. This study aimed to evaluate the role of treadmill exercise in male Wister rats as a single treatment and as an aid-therapy with L-dopa for rotenone-induced PD. To study the role of the Nrf2- ARE pathway as a mechanism involved in exercise-associated improvement in rotenone-induced PD in rats. METHOD Animals were divided into 5 groups, (Control, rotenone, rotenone\exercise, rotenone\L-dopa, and rotenone\exercise\L-dopa (combination)groups). After the PD induction, rats in the rotenone\exercise and combination groups were daily treadmill exercised for 4 weeks. RESULTS Treadmill exercise significantly improved behavioral and motor aspects of rotenone-induced PD. When treadmill exercise was introduced as a single intervention, it amended most behavioral aspects of PD, gait fully corrected, short-term memory, and motor coordination. Where L-dopa corrected locomotor activity and motor coordination but failed to improve short-term memory and only partially corrected the gait of rotenone-treated rats. When treadmill exercise was combined with L-dopa, all features of PD were corrected. It was found that exercise upregulated some of its associative genes to Nrf2 pathways such as TFAM, Nrf2 and NQO.1 mRNA expression. CONCLUSION This study suggests that forced exercise improved parkinsonian like features by activating the Nrf2 pathway.
Collapse
Affiliation(s)
- Dina M Monir
- Department of Physiology, Faculty of Medicine, Sohag University, Sohag, 82524, Egypt
| | - Motamed E Mahmoud
- Department of Animal Behavior and Husbandry (Genetics, Breeding, and Production), Faculty of Veterinary Medicine, Sohag University, Sohag, 82524, Egypt.
| | - Omyma G Ahmed
- Department of Physiology, Faculty of Medicine, Assiut University, Assiut, 71526, Egypt
| | - Ibrahim F Rehan
- Department of Husbandry and Development of Animal Wealth, Faculty of Veterinary Medicine, Menofia University, Shebin Alkom, Menofia, 32511, Egypt
| | - Amany Abdelrahman
- Department of Physiology, Faculty of Medicine, Sohag University, Sohag, 82524, Egypt.
| |
Collapse
|
34
|
Ghoneim FM, Alrefai H, Elsamanoudy AZ, Abo El-khair SM, Khalaf HA. The Protective Role of Prenatal Alpha Lipoic Acid Supplementation against Pancreatic Oxidative Damage in Offspring of Valproic Acid-Treated Rats: Histological and Molecular Study. BIOLOGY 2020; 9:biology9090239. [PMID: 32825436 PMCID: PMC7564314 DOI: 10.3390/biology9090239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 01/21/2023]
Abstract
Background: Sodium valproate (VPA) is an antiepileptic drug (AED) licensed for epilepsy and used during pregnancy in various indications. Alpha-lipoic acid (ALA) is a natural compound inducing endogenous antioxidant production. Our study aimed to investigate the effect of prenatal administration of VPA on the pancreas of rat offspring and assess the potential protective role of ALA co-administration during pregnancy. Methods: Twenty-eight pregnant female albino rats were divided into four groups: group I (negative control), group II (positive control, ALA treated), group III (VPA-treated), and group IV (VPA-ALA-treated). The pancreases of the rat offspring were removed at the fourth week postpartum and prepared for histological, immune-histochemical, morphometric, molecular, and oxidative stress marker studies. Results: In group III, there were pyknotic nuclei, vacuolated cytoplasm with ballooning of acinar, α, and β cells of the pancreas. Ultrastructural degeneration of cytoplasmic organelles was detected. Additionally, there was a significant increase in oxidative stress, a decrease in insulin-positive cell percentage, and an increase in glucagon positive cells in comparison to control groups. Moreover, VPA increased the gene expression of an apoptotic marker, caspase-3, with a decrease in anti-apoptotic Bcl2 and nuclear factor erythroid 2-related factor 2 (Nrf2) transcriptional factor. Conversely, ALA improved oxidative stress and apoptosis in group VI, and a consequent improvement of the histological and ultrastructure picture was detected. Conclusion: ALA co-administration with VPA significantly improved the oxidative stress condition, histological and morphometric picture of the pancreas, and restored normal expression of related genes, including Nrf2, caspase-3, and Bcl-2. Administration of α-lipoic acid has a protective effect against VPA-induced pancreatic oxidative damage via its cytoprotective antioxidant effect.
Collapse
Affiliation(s)
- Fatma M. Ghoneim
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (F.M.G.); (H.A.K.)
| | - Hani Alrefai
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (A.Z.E.); (S.M.A.E.-k.)
- Department of Internal Medicine, Infectious Diseases Div., College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Correspondence: ; Tel.: +1-513-975-9195
| | - Ayman Z. Elsamanoudy
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (A.Z.E.); (S.M.A.E.-k.)
- Clinical Biochemistry Department, Faculty of Medicine, King Abdulaziz University, Jeddah 21465, Saudi Arabia
| | - Salwa M. Abo El-khair
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (A.Z.E.); (S.M.A.E.-k.)
| | - Hanaa A. Khalaf
- Histology and Cell Biology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (F.M.G.); (H.A.K.)
| |
Collapse
|
35
|
Barbosa J, Faria J, Garcez F, Leal S, Afonso LP, Nascimento AV, Moreira R, Queirós O, Carvalho F, Dinis-Oliveira RJ. Repeated Administration of Clinical Doses of Tramadol and Tapentadol Causes Hepato- and Nephrotoxic Effects in Wistar Rats. Pharmaceuticals (Basel) 2020; 13:149. [PMID: 32664348 PMCID: PMC7407499 DOI: 10.3390/ph13070149] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/18/2022] Open
Abstract
Tramadol and tapentadol are fully synthetic and extensively used analgesic opioids, presenting enhanced therapeutic and safety profiles as compared with their peers. However, reports of adverse reactions, intoxications and fatalities have been increasing. Information regarding the molecular, biochemical, and histological alterations underlying their toxicological potential is missing, particularly for tapentadol, owing to its more recent market authorization. Considering the paramount importance of liver and kidney for the metabolism and excretion of both opioids, these organs are especially susceptible to toxicological damage. In the present study, we aimed to characterize the putative hepatic and renal deleterious effects of repeated exposure to therapeutic doses of tramadol and tapentadol, using an in vivo animal model. Male Wistar rats were randomly divided into six experimental groups, composed of six animals each, which received daily single intraperitoneal injections of 10, 25 or 50 mg/kg tramadol or tapentadol (a low, standard analgesic dose, an intermediate dose and the maximum recommended daily dose, respectively). An additional control group was injected with normal saline. Following 14 consecutive days of administration, serum, urine and liver and kidney tissue samples were processed for biochemical, metabolic and histological analysis. Repeated administration of therapeutic doses of both opioids led to: (i) increased lipid and protein oxidation in liver and kidney, as well as to decreased total liver antioxidant capacity; (ii) decreased serum albumin, urea, butyrylcholinesterase and complement C3 and C4 levels, denoting liver synthesis impairment; (iii) elevated serum activity of liver enzymes, such as alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase and γ-glutamyl transpeptidase, as well as lipid profile alterations, also reflecting hepatobiliary commitment; (iv) derangement of iron metabolism, as shown through increases in serum iron, ferritin, haptoglobin and heme oxygenase-1 levels. In turn, elevated serum cystatin C, decreased urine creatinine output and increased urine microalbumin levels were detected upon exposure to tapentadol only, while increased serum amylase and urine N-acetyl-β-D-glucosaminidase activities were observed for both opioids. Collectively, these results are compatible with kidney injury. Changes were also found in the expression levels of liver- and kidney-specific toxicity biomarker genes, upon exposure to tramadol and tapentadol, correlating well with alterations in lipid profile, iron metabolism and glomerular and tubular function. Histopathological analysis evidenced sinusoidal dilatation, microsteatosis, mononuclear cell infiltrates, glomerular and tubular disorganization, and increased Bowman's spaces. Although some findings are more pronounced upon tapentadol exposure, our study shows that, when compared with acute exposure, prolonged administration of both opioids smooths the differences between their toxicological effects, and that these occur at lower doses within the therapeutic range.
Collapse
Affiliation(s)
- Joana Barbosa
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
- UCIBIO, REQUIMTE—Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Juliana Faria
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
- UCIBIO, REQUIMTE—Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
| | - Fernanda Garcez
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
| | - Sandra Leal
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
- Department of Biomedicine, Unit of Anatomy, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- CINTESIS—Center for Health Technology and Services Research, Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Luís Pedro Afonso
- Department of Pathology, Portuguese Institute of Oncology of Porto, 4200-072 Porto, Portugal;
| | - Ana Vanessa Nascimento
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
| | - Roxana Moreira
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
| | - Odília Queirós
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
| | - Félix Carvalho
- UCIBIO, REQUIMTE—Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
| | - Ricardo Jorge Dinis-Oliveira
- IINFACTS—Institute of Research and Advanced Training in Health Sciences and Technologies, Department of Sciences, University Institute of Health Sciences (IUCS), CESPU, CRL, 4585-116 Gandra, Portugal; (J.F.); (F.G.); (S.L.); (A.V.N.); (R.M.); (O.Q.)
- UCIBIO, REQUIMTE—Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
36
|
Abed MN, Alassaf FA, Jasim MHM, Alfahad M, Qazzaz ME. Comparison of Antioxidant Effects of the Proton Pump-Inhibiting Drugs Omeprazole, Esomeprazole, Lansoprazole, Pantoprazole, and Rabeprazole. Pharmacology 2020; 105:645-651. [PMID: 32289807 DOI: 10.1159/000506232] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/26/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Peptic lesions usually develop when there is an imbalance between aggressive drivers and gastro-protective mediators that guard the lining of the gastrointestinal tract. The most crucial of these mediators are antioxidants, whose loss may predispose to oxidative stress, which is believed to be the main aggravator of several diseases including peptic ulcer. Proton pump inhibitors (PPIs) are drugs that are highly effective and widely used for therapeutic management of peptic disorders through inhibition of gastric acid secretion. In spite of this, oxidative damage may continue to be a major issue that can predispose to future lesions. OBJECTIVE The present study is designed to explore the possible antioxidant capability of different PPIs, including omeprazole, esomeprazole, lansoprazole, pantoprazole, and rabeprazole, in an aim to suggest an agent that, in addition to its acid-suppression properties, can provide antioxidant profit. METHODS The antioxidant activity of different PPIs was evaluated calorimetrically to test the ability of each drug to quench oxygen free radical, using the well-known stable free radical α,α-diphenyl-β-picrylhydrazyl (DPPH), and compared to ascorbic acid (AA; vitamin C). The measurements were performed using a spectrophotometer at 517 nm. RESULTS All the studied drugs reduced DPPH, but to different extents. However, omeprazole and esomeprazole showed the highest ability to scavenge free radicals (50% inhibitory concentrations [IC50s] of the percentage for free radical scavenging activity are 18.7 ± 5.7 and 18.7 ± 5.7, respectively, and the AA equivalents are 83,772 ± 11,887 and 81,732 ± 8,523 mg AA/100 g, respectively). Conversely, lansoprazole, pantoprazole, and rabeprazole might be having no role in this story (IC50s of the percentage for free radical scavenging activity are 49.3 ± 3.1, 49 ± 9.4, and 40.7 ± 7.2, respectively, and the AA equivalents are 30,458 ± 3,884, 32,222 ± 10,377, and 37,876 ± 8,816 mg AA/100 g, respectively). CONCLUSION Thus, omeprazole and esomeprazole may confer a significant dual action in gastrointestinal protection by providing potent antioxidant properties in addition to their major role as acid-suppression agents. However, further studies are essential to elucidate the mechanism behind the difference between the drugs of the same class.
Collapse
Affiliation(s)
- Mohammed N Abed
- College of Pharmacy, University of Mosul, Mosul, Iraq, .,Department of Pharmaceutical Chemistry, University of Mosul, Mosul, Iraq,
| | - Fawaz A Alassaf
- College of Pharmacy, University of Mosul, Mosul, Iraq.,Department of Pharmacology and Toxicology, University of Mosul, Mosul, Iraq
| | - Mahmood H M Jasim
- College of Pharmacy, University of Mosul, Mosul, Iraq.,Department of Pharmaceutical Chemistry, University of Mosul, Mosul, Iraq
| | - Mohanad Alfahad
- College of Pharmacy, University of Mosul, Mosul, Iraq.,Department of Pharmaceutics, University of Mosul, Mosul, Iraq
| | - Mohannad E Qazzaz
- College of Pharmacy, University of Mosul, Mosul, Iraq.,Department of Pharmacognosy and Medicinal Plants, University of Mosul, Mosul, Iraq
| |
Collapse
|
37
|
Nazmeen A, Chen G, Ghosh TK, Maiti S. Breast cancer pathogenesis is linked to the intra-tumoral estrogen sulfotransferase (hSULT1E1) expressions regulated by cellular redox dependent Nrf-2/NF κβ interplay. Cancer Cell Int 2020; 20:70. [PMID: 32158360 PMCID: PMC7057506 DOI: 10.1186/s12935-020-1153-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Estrogen sulfotransferase catalyzes conjugation of sulfuryl-group to estradiol/estrone and regulates E2 availability/activity via estrogen-receptor or non-receptor mediated pathways. Sulfoconjugated estrogen fails to bind estrogen-receptor (ER). High estrogen is a known carcinogen in postmenopausal women. Reports reveal a potential redox-regulation of hSULT1E1/E2-signalling. Further, oxidatively-regulated nuclear-receptor-factor 2 (Nrf2) and NFκβ in relation to hSULT1E1/E2 could be therapeutic-target via cellular redox-modification. METHODS Here, oxidative stress-regulated SULT1E1-expression was analyzed in human breast carcinoma-tissues and in rat xenografted with human breast-tumor. Tumor and its surrounding tissues were obtained from the district-hospital. Intracellular redox-environment of tumors was screened with some in vitro studies. RT-PCR and western blotting was done for SULT1E1 expression. Immunohistochemistry was performed to analyze SULT1E1/Nrf2/NFκβ localization. Tissue-histoarchitecture/DNA-stability (comet assay) studies were done. RESULTS Oxidative-stress induces SULT1E1 via Nrf2/NFκβ cooperatively in tumor-pathogenesis to maintain the required proliferative-state under enriched E2-environment. Higher malondialdehyde/non-protein-soluble-thiol with increased superoxide-dismutase/glutathione-peroxidase/catalase activities was noticed. SULT1E1 expression and E2-level were increased in tumor-tissue compared to their corresponding surrounding-tissues. CONCLUSIONS It may be concluded that tumors maintain a sustainable oxidative-stress through impaired antioxidants as compared to the surrounding. Liver-tissues from xenografted rat manifested similar E2/antioxidant dysregulations favoring pre-tumorogenic environment.
Collapse
Affiliation(s)
- Aarifa Nazmeen
- Dept. of Biochemistry, Cell & Molecular Therapeutics Lab, Oriental Institute of Science & Technology, Midnapore, 721101 India
| | - Guangping Chen
- Venture I OSU Laboratory, Oklahoma Technology & Research Park, 1110 S. Innovation Way, Stillwater, OK 74074 USA
| | - Tamal Kanti Ghosh
- Special Secretary, Higher Medical Education, Health and Family Welfare Dept, Govt. of West Bengal, Salt Lake, Calcutta, India
| | - Smarajit Maiti
- Dept. of Biochemistry, Cell & Molecular Therapeutics Lab, Oriental Institute of Science & Technology, Midnapore, 721101 India
- Department of Biochemistry and Biotechnology, Cell & Molecular Therapeutics Lab, OIST, Midnapore, 721102 India
| |
Collapse
|
38
|
Rizk NI, Rizk MS, Mohamed AS, Naguib YM. Attenuation of sleep deprivation dependent deterioration in male fertility parameters by vitamin C. Reprod Biol Endocrinol 2020; 18:2. [PMID: 31926565 PMCID: PMC6954532 DOI: 10.1186/s12958-020-0563-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/06/2020] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Male fertility is multifaceted and its integrity is as well multifactorial. Normal spermatogenesis is dependent on competent testicular function; namely normal anatomy, histology, physiology and hormonal regulation. Lifestyle stressors, including sleep interruption and even deprivation, have been shown to seriously impact male fertility. We studied here both the effects and the possible underlying mechanisms of vitamin C on male fertility in sleep deprived rats. METHODS Thirty male Wistar albino rats were used in the present study. Rats were divided (10/group) into: control (remained in their cages with free access to food and water), sleep deprivation (SD) group (subjected to paradoxical sleep deprivation for 5 consequent days, rats received intra-peritoneal injections of vehicle daily throughout the sleep deprivation), and sleep deprivation vitamin C-treated (SDC) group (subjected to sleep deprivation for 5 consequent days with concomitant intra-peritoneal injections of 100 mg/kg/day vitamin C). Sperm analysis, hormonal assay, and measurement of serum oxidative stress and inflammatory markers were performed. Testicular gene expression of Nrf2 and NF-κβ was assessed. Structural changes were evaluated by testicular histopathology, while PCNA immunostaining was conducted to assess spermatogenesis. RESULTS Sleep deprivation had significantly altered sperm motility, viability, morphology and count. Serum levels of cortisol, corticosterone, IL-6, IL-17, MDA were increased, while testosterone and TAC levels were decreased. Testicular gene expression of Nrf2 was decreased, while NF-κβ was increased. Sleep deprivation caused structural changes in the testes, and PCNA immunostaining showed defective spermatogenesis. Administration of vitamin C significantly countered sleep deprivation induced deterioration in male fertility parameters. CONCLUSION Treatment with vitamin C enhanced booth testicular structure and function in sleep deprived rats. Vitamin C could be a potential fertility enhancer against lifestyle stressors.
Collapse
Affiliation(s)
- Nermin I Rizk
- Clinical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Mohamed S Rizk
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Asmaa S Mohamed
- Pathology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt
| | - Yahya M Naguib
- Clinical Physiology Department, Faculty of Medicine, Menoufia University, Menoufia, Egypt.
| |
Collapse
|
39
|
Nakatake R, Hishikawa H, Kotsuka M, Ishizaki M, Matsui K, Nishizawa M, Yoshizawa K, Kaibori M, Okumura T. The Proton Pump Inhibitor Lansoprazole Has Hepatoprotective Effects in In Vitro and In Vivo Rat Models of Acute Liver Injury. Dig Dis Sci 2019; 64:2854-2866. [PMID: 30989463 DOI: 10.1007/s10620-019-05622-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 04/08/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND/AIMS The proton pump inhibitor lansoprazole (LPZ) is clinically used to reduce gastric acid secretion, but little is known about its possible hepatoprotective effects. This study aimed to investigate the hepatoprotective effects of LPZ and its potential mechanisms using in vitro and in vivo rat models of liver injury. METHODS For the in vitro model of liver injury, primary cultured rat hepatocytes were treated with interleukin-1β in the presence or absence of LPZ. The influence of LPZ on inducible nitric oxide synthase (iNOS) induction and nitric oxide (NO) production and on the associated signaling pathways was analyzed. For the in vivo model, rats were treated with D-galactosamine (GalN) and lipopolysaccharide (LPS). The effects of LPZ on survival and proinflammatory mediator expression (including iNOS and tumor necrosis factor-α) in these rats were examined. RESULTS LPZ inhibited iNOS induction partially through suppression of the nuclear factor-kappa B signaling pathway in hepatocytes, thereby reducing potential liver injury from excessive NO levels. Additionally, LPZ increased survival by 50% and decreased iNOS, tumor necrosis factor-α, and cytokine-induced neutrophil chemoattractant-1 mRNA expression in the livers of GalN/LPS-treated rats. LPZ also inhibited nuclear factor-kappa B activation by GalN/LPS. CONCLUSIONS LPZ inhibits the induction of several inflammatory mediators (including cytokines, chemokines, and NO) partially through suppression of nuclear factor-kappa B, resulting in the prevention of fulminant liver failure. The therapeutic potential of LPZ for liver injuries warrants further investigation.
Collapse
Affiliation(s)
- Richi Nakatake
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.
| | - Hidehiko Hishikawa
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Masaya Kotsuka
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Morihiko Ishizaki
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Kosuke Matsui
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Mikio Nishizawa
- Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| | - Katsuhiko Yoshizawa
- Laboratory of Environmental Sciences, Department of Food Sciences and Nutrition, School of Human Environmental Sciences, Mukogawa Women's University, 6-46 Ikebiraki-cho, Nishinomiya, Hyogo, 663-8558, Japan
| | - Masaki Kaibori
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan
| | - Tadayoshi Okumura
- Department of Surgery, Kansai Medical University, 2-5-1 Shinmachi, Hirakata, Osaka, 573-1010, Japan.,Research Organization of Science and Technology, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan
| |
Collapse
|
40
|
Hammond CL, Roztocil E, Phipps RP, Feldon SE, Woeller CF. Proton pump inhibitors attenuate myofibroblast formation associated with thyroid eye disease through the aryl hydrocarbon receptor. PLoS One 2019; 14:e0222779. [PMID: 31536596 PMCID: PMC6752849 DOI: 10.1371/journal.pone.0222779] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/06/2019] [Indexed: 12/15/2022] Open
Abstract
Thyroid eye disease (TED) can lead to scar formation and tissue remodeling in the orbital space. In severe cases, the scarring process leads to sight-threatening pathophysiology. There is no known effective way to prevent scar formation in TED patients, or to reverse scarring once it occurs. In this study, we show that the proton pump inhibitors (PPIs), esomeprazole and lansoprazole, can prevent transforming growth factor beta (TGFβ)-mediated differentiation of TED orbital fibroblasts to myofibroblasts, a critical step in scar formation. Both PPIs prevent TGFβ-induced increases in alpha-smooth muscle actin (αSMA), calponin, and collagen production and reduce TED orbital fibroblast cell proliferation and migration. Esomeprazole and lansoprazole exert these effects through an aryl hydrocarbon receptor (AHR)-dependent pathway that includes reducing β-catenin/Wnt signaling. We conclude that PPIs are potentially useful therapies for preventing or treating TED by reducing the myofibroblast accumulation that occurs in the disease.
Collapse
Affiliation(s)
- Christine L. Hammond
- Flaum Eye Institute, School of Medicine and Dentistry, University of Rochester, Rochester, New York, United States of America
| | - Elisa Roztocil
- Flaum Eye Institute, School of Medicine and Dentistry, University of Rochester, Rochester, New York, United States of America
| | - Richard P. Phipps
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, New York, United States of America
| | - Steven E. Feldon
- Flaum Eye Institute, School of Medicine and Dentistry, University of Rochester, Rochester, New York, United States of America
| | - Collynn F. Woeller
- Flaum Eye Institute, School of Medicine and Dentistry, University of Rochester, Rochester, New York, United States of America
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, New York, United States of America
| |
Collapse
|
41
|
Hafez HM, Ibrahim MA, Zedan MZ, Hassan M, Hassanein H. Nephroprotective effect of cilostazol and verapamil against thioacetamide-induced toxicity in rats may involve Nrf2/HO-1/NQO-1 signaling pathway. Toxicol Mech Methods 2018; 29:146-152. [DOI: 10.1080/15376516.2018.1528648] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Heba M. Hafez
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Mohamed A. Ibrahim
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Mervat Z. Zedan
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Marwa Hassan
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Hanaa Hassanein
- Department of Histology, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
42
|
Hussein RM, Mohamed WR, Omar HA. A neuroprotective role of kaempferol against chlorpyrifos-induced oxidative stress and memory deficits in rats via GSK3β-Nrf2 signaling pathway. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2018; 152:29-37. [PMID: 30497708 DOI: 10.1016/j.pestbp.2018.08.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 06/25/2018] [Accepted: 08/10/2018] [Indexed: 06/09/2023]
Abstract
Chlorpyrifos (CPF) is an agricultural pesticide and a potential food contaminant, which causes neurotoxicity. Here, we aimed at exploring the link between the repeated exposure to CPF and memory dysfunction in rats and the possible protective effect of kaempferol, a flavonoid with appreciable antioxidant and anti-inflammatory activities. Rats were divided into: Control group (received drug vehicles for 14 days); CPF-treated group (received subcutaneous 18 mg/kg BW of CPF daily for 14 days and CPF + Kaempferol treated group (received the same CPF dose +21 mg/kg BW of Kaempferol intraperitoneally for 14 days. On the 14th day, Y-maze and novel target recognition behavioral tests were employed to evaluate memory deficits. 24 h after the last dose of CPF, animals were sacrificed, and brain tissues were used for the determination of oxidative stress biomarkers and gene expression levels of GSK3β and Nrf2. The results revealed that CPF-treated rats suffered from severe deterioration of spatial and non-spatial memory functions with low activities of antioxidant enzymes and acetylcholinesterase (AChE). The administration of kaempferol significantly protected against CPF-induced neuronal damage, increased the activities of antioxidant enzymes and AChE and induced a better performance in the behavioral tests. The protective effect of kaempferol was mediated through the inhibition of GSK3β gene expression and the induction of Nrf2 expression in the brain tissues. In conclusion, the repeated exposure to CPF is associated with oxidative stress and memory deficits in rats. However, kaempferol administration effectively alleviated CPF- induced brain toxicity, possibly through the modulation of GSK3β-Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Rasha M Hussein
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, 62514 Beni-Suef, Egypt.
| | - Wafaa R Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| | - Hany A Omar
- Sharjah Institute for Medical Research and College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
43
|
Selim SA, El-Baset SAA, Kattaia AAA, Askar EM, Elkader EA. Bone marrow-derived mesenchymal stem cells ameliorate liver injury in a rat model of sepsis by activating Nrf2 signaling. Histochem Cell Biol 2018; 151:249-262. [PMID: 30250973 DOI: 10.1007/s00418-018-1731-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2018] [Indexed: 01/08/2023]
Abstract
Sepsis is a fatal condition that leads to serious systemic inflammation and multiple organ dysfunction syndromes. This study was designed to investigate the possible therapeutic effect of bone marrow-derived mesenchymal stem cells (BMSCs) on sepsis-induced liver injury. We also aimed to examine the role of Nrf2 activation in modulating the response to sepsis following BMSCs treatment. Twenty-four adult male albino rats were assigned to: control, lipopolysaccharide (LPS) and LPS-stem cell groups. Liver samples were processed for light and electron microscope examinations. Immunohistochemical localization of BAX, proliferating cell nuclear antigen and nuclear factor-erythroid 2-related factor 2 (Nrf2) was carried out. Liver homogenates were prepared for assessment of reduced glutathione, glutathione peroxidase, tumor necrosis factor-alpha and interleukin-6 and also real-time PCR analysis of Nrf2 expression. BMSCs treatment improved the histopathological changes of the liver, enhanced tissue regeneration and decreased apoptosis following sepsis. We reported highly significant enhancement in Nrf2 expressions at mRNA and protein levels in the LPS-stem cell group compared with the LPS group. The up regulation of Nrf2 was probably implicated in decreasing inflammatory cytokine levels and counteracting oxidative stress induced by sepsis. Thus, BMSCs therapies could be a viable approach to treat sepsis-induced liver damage by activating Nrf2 signaling.
Collapse
Affiliation(s)
- Sally A Selim
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Koliat Al Tob Street, Zagazig, Ash Sharqia Governorate, 44519, Egypt
| | - Samia A Abd El-Baset
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Koliat Al Tob Street, Zagazig, Ash Sharqia Governorate, 44519, Egypt
| | - Asmaa A A Kattaia
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Koliat Al Tob Street, Zagazig, Ash Sharqia Governorate, 44519, Egypt.
| | - Eman M Askar
- Department of Histology and Cell Biology, Faculty of Medicine, Zagazig University, Koliat Al Tob Street, Zagazig, Ash Sharqia Governorate, 44519, Egypt
| | - Eman Abd Elkader
- Department of Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
44
|
Abstract
The NFE2L2 gene encodes the transcription factor Nrf2 best known for regulating the expression of antioxidant and detoxification genes. Gene knockout approaches have demonstrated its universal cytoprotective features. While Nrf2 has been the topic of intensive research in cancer biology since its discovery in 1994, understanding the role of Nrf2 in cardiovascular disease has just begun. The literature concerning Nrf2 in experimental models of atherosclerosis, ischemia, reperfusion, cardiac hypertrophy, heart failure, and diabetes supports its cardiac protective character. In addition to antioxidant and detoxification genes, Nrf2 has been found to regulate genes participating in cell signaling, transcription, anabolic metabolism, autophagy, cell proliferation, extracellular matrix remodeling, and organ development, suggesting that Nrf2 governs damage resistance as well as wound repair and tissue remodeling. A long list of small molecules, most derived from natural products, have been characterized as Nrf2 inducers. These compounds disrupt Keap1-mediated Nrf2 ubquitination, thereby prohibiting proteasomal degradation and allowing Nrf2 protein to accumulate and translocate to the nucleus, where Nrf2 interacts with sMaf to bind to ARE in the promoter of genes. Recently alternative mechanisms driving Nrf2 protein increase have been revealed, including removal of Keap1 by autophagy due to p62/SQSTM1 binding, inhibition of βTrCP or Synoviolin/Hrd1-mediated ubiquitination of Nrf2, and de novo Nrf2 protein translation. We review here a large volume of literature reporting historical and recent discoveries about the function and regulation of Nrf2 gene. Multiple lines of evidence presented here support the potential of dialing up the Nrf2 pathway for cardiac protection in the clinic.
Collapse
Affiliation(s)
- Qin M Chen
- Department of Pharmacology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Anthony J Maltagliati
- Department of Pharmacology, College of Medicine, University of Arizona , Tucson, Arizona
| |
Collapse
|
45
|
Nishi T, Yamamoto Y, Yamagishi N, Iguchi M, Tamai H, Ito T, Tsuruo Y, Ichinose M, Kitano M, Ueyama T. Lansoprazole prevents the progression of liver fibrosis in non-alcoholic steatohepatitis model rats. ACTA ACUST UNITED AC 2018; 70:383-392. [PMID: 29355950 DOI: 10.1111/jphp.12870] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/25/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVES We previously demonstrated that lansoprazole provided hepatoprotection in a drug-induced hepatitis animal model partially through the Nrf2/HO-1 pathway. Here, we examined whether lansoprazole could also provide hepatoprotection in a rat model of non-alcoholic steatohepatitis (NASH). METHODS Six-week-old rats were fed a normal chow or a choline-deficient amino acid-defined (CDAA) diet to establish a rat model of NASH. The groups fed a CDAA diet for 5 weeks were subcutaneously administered either a vehicle or a lansoprazole suspension for 4 weeks beginning the second week of the experiment. KEY FINDINGS Bridging fibrosis was observed in the livers of almost all the NASH model rats (six of seven), but it was not always observed in NASH model rats (one of seven) continuously administered lansoprazole. The serum aspartate aminotransferase level elevated by the CDAA diet was significantly decreased following lansoprazole administration. Lansoprazole also increased the expression of Nrf2, but not HO-1, in the liver of NASH model rats. Lansoprazole decreased the level of activated TGF-β protein. Furthermore, interleukin-6 gene and protein expression were decreased. CONCLUSIONS Lansoprazole inhibits hepatic fibrogenesis, at least during the early stages, in CDAA diet-induced NASH model rats. The mechanisms might be associated with cytokine suppression but not the inhibition of reactive oxygen species.
Collapse
Affiliation(s)
- Toshio Nishi
- Department of Anatomy and Cell Biology, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Yuta Yamamoto
- Department of Anatomy and Cell Biology, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Naoko Yamagishi
- Department of Anatomy and Cell Biology, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Mikitaka Iguchi
- 2nd Department of Internal Medicine, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Hideyuki Tamai
- 2nd Department of Internal Medicine, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Takao Ito
- Department of Anatomy and Cell Biology, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Yoshihiro Tsuruo
- Department of Anatomy and Cell Biology, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan.,Department of Anatomy and Cell Biology, Tokushima University Graduate School of Medical Science, Tokushima, Japan
| | - Masao Ichinose
- 2nd Department of Internal Medicine, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Masayuki Kitano
- 2nd Department of Internal Medicine, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| | - Takashi Ueyama
- Department of Anatomy and Cell Biology, Wakayama Medical University Graduate School of Medicine, Wakayama, Japan
| |
Collapse
|
46
|
Drug Repurposing Patent Applications July–September 2017. Assay Drug Dev Technol 2017; 15:378-382. [DOI: 10.1089/adt.2017.29069.pq3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
47
|
Khaleel SA, Alzokaky AA, Raslan NA, Alwakeel AI, Abd El-Aziz HG, Abd-Allah AR. Lansoprazole halts contrast induced nephropathy through activation of Nrf2 pathway in rats. Chem Biol Interact 2017; 270:33-40. [DOI: 10.1016/j.cbi.2017.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 04/12/2017] [Indexed: 12/11/2022]
|
48
|
Lo JY, Spatola BN, Curran SP. WDR23 regulates NRF2 independently of KEAP1. PLoS Genet 2017; 13:e1006762. [PMID: 28453520 PMCID: PMC5428976 DOI: 10.1371/journal.pgen.1006762] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 05/12/2017] [Accepted: 04/13/2017] [Indexed: 12/22/2022] Open
Abstract
Cellular adaptation to stress is essential to ensure organismal survival. NRF2/NFE2L2 is a key determinant of xenobiotic stress responses, and loss of negative regulation by the KEAP1-CUL3 proteasome system is implicated in several chemo- and radiation-resistant cancers. Advantageously using C. elegans alongside human cell culture models, we establish a new WDR23-DDB1-CUL4 regulatory axis for NRF2 activity that operates independently of the canonical KEAP1-CUL3 system. WDR23 binds the DIDLID sequence within the Neh2 domain of NRF2 to regulate its stability; this regulation is not dependent on the KEAP1-binding DLG or ETGE motifs. The C-terminal domain of WDR23 is highly conserved and involved in regulation of NRF2 by the DDB1-CUL4 complex. The addition of WDR23 increases cellular sensitivity to cytotoxic chemotherapeutic drugs and suppresses NRF2 in KEAP1-negative cancer cell lines. Together, our results identify WDR23 as an alternative regulator of NRF2 proteostasis and uncover a cellular pathway that regulates NRF2 activity and capacity for cytoprotection independently of KEAP1.
Collapse
Affiliation(s)
- Jacqueline Y. Lo
- University of Southern California, Leonard Davis School of Gerontology, Los Angeles, California, United States of America
- University of Southern California, Dornsife College of Letters, Arts, and Sciences, Department of Molecular and Computational Biology, Los Angeles, California, United States of America
| | - Brett N. Spatola
- University of Southern California, Leonard Davis School of Gerontology, Los Angeles, California, United States of America
- University of Southern California, Dornsife College of Letters, Arts, and Sciences, Department of Molecular and Computational Biology, Los Angeles, California, United States of America
| | - Sean P. Curran
- University of Southern California, Leonard Davis School of Gerontology, Los Angeles, California, United States of America
- University of Southern California, Dornsife College of Letters, Arts, and Sciences, Department of Molecular and Computational Biology, Los Angeles, California, United States of America
| |
Collapse
|
49
|
Dai C, Ciccotosto GD, Cappai R, Tang S, Li D, Xie S, Xiao X, Velkov T. Curcumin Attenuates Colistin-Induced Neurotoxicity in N2a Cells via Anti-inflammatory Activity, Suppression of Oxidative Stress, and Apoptosis. Mol Neurobiol 2016; 55:421-434. [PMID: 27957686 DOI: 10.1007/s12035-016-0276-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/30/2016] [Indexed: 01/06/2023]
Abstract
Neurotoxicity is an unwanted side-effect seen in patients receiving therapy with the "last-line" polymyxin antibiotics. This is the first study to show that colistin-induced neurotoxicity in neuroblastoma-2a (N2a) cells gives rise to an inflammatory response involving the IL-1β/p-IκB-α/NF-κB pathway. Pretreatment with curcumin at 5, 10, and 20 μM for 2 h prior to colistin (200 μM) exposure for 24 h, produced an anti-inflammatory effect by significantly down-regulating the expression of the pro-inflammatory mediators cyclooxygenase-2 (COX-2), phosphorylation of the inhibitor of nuclear factor-kappa B (NF-κB) (p-IκB)-α, and concomitantly NF-κB levels. Moreover, curcumin significantly decreased intracellular reactive oxygen species (ROS) production and increased the activities of the anti-ROS enzymes superoxide dismutase, catalase, and the intracellular levels of glutathione. Curcumin pretreatment also protected the cells from colistin-induced mitochondrial dysfunction, caspase activation, and subsequent apoptosis. Overall, our findings demonstrate for the first time, a potential role for curcumin for treating polymyxin-induced neurotoxicity through the modulation of NF-κB signaling and its potent anti-oxidative and anti-apoptotic effects.
Collapse
Affiliation(s)
- Chongshan Dai
- College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Giuseppe D Ciccotosto
- Department of Pathology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Roberto Cappai
- Department of Pathology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Shusheng Tang
- College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Daowen Li
- College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Sanlei Xie
- College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China
| | - Xilong Xiao
- College of Veterinary Medicine, China Agricultural University, 2 Yuanmingyuan West Road, Beijing, 100193, People's Republic of China.
| | - Tony Velkov
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
50
|
Dai C, Li B, Zhou Y, Li D, Zhang S, Li H, Xiao X, Tang S. Curcumin attenuates quinocetone induced apoptosis and inflammation via the opposite modulation of Nrf2/HO-1 and NF-kB pathway in human hepatocyte L02 cells. Food Chem Toxicol 2016; 95:52-63. [DOI: 10.1016/j.fct.2016.06.025] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/18/2016] [Accepted: 06/25/2016] [Indexed: 01/06/2023]
|