1
|
Gencer G, Sarikurkcu C, Tepe B. Unveiling the Phytochemical Diversity and Bioactivity of Astragalus melanophrurius: A First Report Integrating Experimental and In Silico Approaches. Pharmaceuticals (Basel) 2025; 18:103. [PMID: 39861165 PMCID: PMC11768182 DOI: 10.3390/ph18010103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Background: The genus Astragalus is renowned for its diverse bioactive potential, yet the chemical composition and biological properties of Astragalus melanophrurius remain inadequately explored. This study aimed to investigate the chemical profile, antioxidant capacity, and enzyme inhibitory activities of methanol extracts from various plant parts of A. melanophrurius. Methods: Methanol extracts were obtained from leaves, stems, flowers, roots, and aerial portions of A. melanophrurius. The chemical composition was determined using LC-ESI-MS/MS, focusing on key phytochemicals such as hyperoside, kaempferol, 4-hydroxybenzoic acid, and chlorogenic acid. Antioxidant activities were assessed via DPPH, ABTS, and FRAP assays, while enzyme inhibitory activities were evaluated against α-amylase and tyrosinase. In silico molecular docking analyses were conducted to explore the interactions between major compounds and target enzymes. Results: The leaf extract exhibited the highest total phenolic and flavonoid contents, correlating with superior antioxidant activities, achieving IC50 values of 16.55 mg/mL, 4.58 mg/mL, and 3.07 mg/mL in DPPH, ABTS, and FRAP assays, respectively. The root extract demonstrated notable α-amylase (IC50 = 2.99 mg/mL) and tyrosinase (IC50 = 1.34 mg/mL) inhibitory activities, suggesting potential applications in diabetes and hyperpigmentation management. Molecular docking revealed stable complexes of hyperoside and kaempferol with target enzymes, supporting their roles in observed bioactivities. Conclusions: This study highlights the bioactivity of A. melanophrurius extracts, particularly from leaves and roots, supporting their therapeutic potential. Future research should focus on isolating active compounds and conducting in vivo studies to confirm efficacy and elucidate mechanisms of action.
Collapse
Affiliation(s)
- Gulcan Gencer
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Afyonkarahisar Health Sciences University, 03030 Afyonkarahisar, Türkiye
| | - Cengiz Sarikurkcu
- Department of Analytical Chemistry, Faculty of Pharmacy, Afyonkarahisar Health Sciences University, 03100 Afyonkarahisar, Türkiye;
| | - Bektas Tepe
- Department of Molecular Biology and Genetics, Faculty of Science, Kilis 7 Aralik University, 79000 Kilis, Türkiye;
| |
Collapse
|
2
|
Zeng Z, Hu J, Xiao G, Liu Y, Jia D, Wu G, Xie C, Li S, Bi X. Integrating network toxicology and molecular docking to explore the toxicity of the environmental pollutant butyl hydroxyanisole: An example of induction of chronic urticaria. Heliyon 2024; 10:e35409. [PMID: 39170477 PMCID: PMC11336633 DOI: 10.1016/j.heliyon.2024.e35409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
The study aimed to comprehensively investigate environmental pollutants' potential toxicity and underlying molecular mechanisms, focusing on chronic urticaria (CU) induced by butylated hydroxyanisole (BHA) exposure, further drawing public awareness regarding the potential risks of environmental pollutants, applying ChEMBL, STITCH, and SwissTargetPrediction databases to predict the targets of BHA, CTD, GeneCards, and OMIM databases to collect the relevant targets of CU. Ultimately, we identified 81 potential targets of BHA-induced CU and extracted 31 core targets, including TNF, SRC, CASP3, BCL2, IL2, and MMP9. GO and KEGG enrichment analyses revealed that these core targets were predominantly involved in cancer signaling, estrogen and endocrine resistance pathways. Furthermore, molecular docking confirmed the ability of BHA to bind with core targets. The onset and development of CU may result from BHA by affecting multiple immune signaling pathways. Our study elucidated the molecular mechanisms of BHA toxicity and its role in CU induction, providing the basis for preventing and treating chronic urticaria associated with environmental BHA exposure.
Collapse
Affiliation(s)
- Zhihao Zeng
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jiaoting Hu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Guanlin Xiao
- Guangdong Provincial Engineering and Technology Research Institute of Traditional Chinese Medicine/Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China
| | - Yanchang Liu
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Dezheng Jia
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Guangying Wu
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Canhui Xie
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Sumei Li
- Guangdong Provincial Engineering and Technology Research Institute of Traditional Chinese Medicine/Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China
| | - Xiaoli Bi
- Guangdong Provincial Engineering and Technology Research Institute of Traditional Chinese Medicine/Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, 510095, China
| |
Collapse
|
3
|
Choi JY, Byeon HW, Park SO, Uyangaa E, Kim K, Eo SK. Inhibition of NADPH oxidase 2 enhances resistance to viral neuroinflammation by facilitating M1-polarization of macrophages at the extraneural tissues. J Neuroinflammation 2024; 21:115. [PMID: 38698374 PMCID: PMC11067137 DOI: 10.1186/s12974-024-03078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/27/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Macrophages play a pivotal role in the regulation of Japanese encephalitis (JE), a severe neuroinflammation in the central nervous system (CNS) following infection with JE virus (JEV). Macrophages are known for their heterogeneity, polarizing into M1 or M2 phenotypes in the context of various immunopathological diseases. A comprehensive understanding of macrophage polarization and its relevance to JE progression holds significant promise for advancing JE control and therapeutic strategies. METHODS To elucidate the role of NADPH oxidase-derived reactive oxygen species (ROS) in JE progression, we assessed viral load, M1 macrophage accumulation, and cytokine production in WT and NADPH oxidase 2 (NOX2)-deficient mice using murine JE model. Additionally, we employed bone marrow (BM) cell-derived macrophages to delineate ROS-mediated regulation of macrophage polarization by ROS following JEV infection. RESULTS NOX2-deficient mice exhibited increased resistance to JE progression rather than heightened susceptibility, driven by the regulation of macrophage polarization. These mice displayed reduced viral loads in peripheral lymphoid tissues and the CNS, along with diminished infiltration of inflammatory cells into the CNS, thereby resulting in attenuated neuroinflammation. Additionally, NOX2-deficient mice exhibited enhanced JEV-specific Th1 CD4 + and CD8 + T cell responses and increased accumulation of M1 macrophages producing IL-12p40 and iNOS in peripheral lymphoid and inflamed extraneural tissues. Mechanistic investigations revealed that NOX2-deficient macrophages displayed a more pronounced differentiation into M1 phenotypes in response to JEV infection, thereby leading to the suppression of viral replication. Importantly, the administration of H2O2 generated by NOX2 was shown to inhibit M1 macrophage polarization. Finally, oral administration of the ROS scavenger, butylated hydroxyanisole (BHA), bolstered resistance to JE progression and reduced viral loads in both extraneural tissues and the CNS, along with facilitated accumulation of M1 macrophages. CONCLUSION In light of our results, it is suggested that ROS generated by NOX2 play a role in undermining the control of JEV replication within peripheral extraneural tissues, primarily by suppressing M1 macrophage polarization. Subsequently, this leads to an augmentation in the viral load invading the CNS, thereby facilitating JE progression. Hence, our findings ultimately underscore the significance of ROS-mediated macrophage polarization in the context of JE progression initiated JEV infection.
Collapse
Affiliation(s)
- Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Hee Won Byeon
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Seong Ok Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Erdenebileg Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, 54596, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, 54596, Republic of Korea.
| |
Collapse
|
4
|
Mut-Salud N, Guardia JJ, Fernández A, Blancas I, Zentar H, Garrido JM, Álvarez-Manzaneda E, Chahboun R, Rodríguez-Serrano F. Discovery of a synthetic taiwaniaquinoid with potent in vitro and in vivo antitumor activity against breast cancer cells. Biomed Pharmacother 2023; 168:115791. [PMID: 37924793 DOI: 10.1016/j.biopha.2023.115791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/12/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023] Open
Abstract
Taiwaniaquinoids are a unique family of diterpenoids predominantly isolated from Taiwania cryptomerioides Hayata. Previously, we evaluated the antiproliferative effect of several synthetic taiwaniaquinoids against human lung (A-549), colon (T-84), and breast (MCF-7) tumor cell lines. Herein, we report the in vitro and in vivo antitumor activity of the most potent compounds. Their cytotoxic activity against healthy peripheral blood mononuclear cells (PBMCs) has also been examined. We underscore the limited toxicity of compound C36 in PBMCs and demonstrate that it exerts its antitumor effect in MCF-7 cells (IC50 = 1.8 µM) by triggering an increase in reactive oxygen species, increasing the cell population in the sub-G1 phase of the cell cycle (90 %), and ultimately activating apoptotic (49.6 %) rather than autophagic processes. Western blot results suggested that the underlying mechanism of the C36 apoptotic effects was linked to caspase 9 activation and a rise in the Bax/Bcl-2 ratio. In vivo analyses showed normal behavior and hematological parameters in C57BL/6 mice post C36 treatment. Moreover, no significant impact was observed on the biochemical parameters of these animals, indicating that C36 did not induce liver toxicity. Furthermore, C36 demonstrated a significant reduction in tumor growth in immune-competent C57BL/6 mice implanted with E0771 mouse mammary tumor cells, effectively improving survival rates. These findings position taiwaniaquinoids, particularly compound C36, as promising therapeutic candidates for human breast cancer.
Collapse
Affiliation(s)
- Nuria Mut-Salud
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada 18016, Spain
| | - Juan J Guardia
- Department of Organic Chemistry, Faculty of Sciences, University of Granada, Granada 18071, Spain
| | - Antonio Fernández
- Department of Organic Chemistry, Faculty of Sciences, University of Granada, Granada 18071, Spain
| | - Isabel Blancas
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada 18016, Spain; Department of Medicine, School of Medicine, University of Granada, Granada 18016, Spain; Department of Medical Oncology, San Cecilio University Hospital, Granada 18016, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), Granada 18014, Spain
| | - Houda Zentar
- Department of Organic Chemistry, Faculty of Sciences, University of Granada, Granada 18071, Spain
| | - José M Garrido
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada 18016, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), Granada 18014, Spain; Department of Surgery and Surgical Specialties, University of Granada, Granada 18016, Spain
| | | | - Rachid Chahboun
- Department of Organic Chemistry, Faculty of Sciences, University of Granada, Granada 18071, Spain.
| | - Fernando Rodríguez-Serrano
- Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, Granada 18016, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), Granada 18014, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada 18016, Spain.
| |
Collapse
|
5
|
Zhang XJ, Diao MN, Zhang YF. A review of the occurrence, metabolites and health risks of butylated hydroxyanisole (BHA). JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:6150-6166. [PMID: 37127924 DOI: 10.1002/jsfa.12676] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 04/19/2023] [Accepted: 05/02/2023] [Indexed: 05/03/2023]
Abstract
Butylated hydroxyanisole (BHA) is mainly used as a food additive due to its antioxidant properties, which prevent or delay oxidation reactions and extend the storage life of products. The widespread use of BHA has led to its extensive presence in various environmental matrices and human tissues. Food intake is the main route of human exposure to BHA. Under different conditions, BHA can produce different metabolites, with tert-butyl hydroquinone (TBHQ) being one of the major products. Several studies have shown that BHA could cause thyroid system damage, metabolic and growth disorders, neurotoxicity, and carcinogenesis. Mechanisms such as endocrine disruption, genotoxicity, disturbances of energy metabolism, reactive oxygen species (ROS) production, signaling pathways, and imbalances in calcium homeostasis appear to be associated with the toxic effects of BHA. Avoiding the toxic effects of BHA to the maximum extent possible is a top priority. Finding safe, non-toxic and environmentally friendly alternatives to BHA should be the focus of subsequent research. In all, this review summarized the current situation related to BHA and might make recommendations for future research directions. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiao-Jing Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Mei-Ning Diao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yin-Feng Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Eltayeb HA, Stewart L, Morgem M, Johnson T, Nguyen M, Earl K, Sodipe A, Jackson D, Olufemi SE. Antioxidants Amelioration Is Insufficient to Prevent Acrylamide and Alpha-Solanine Synergistic Toxicity in BEAS-2B Cells. Int J Mol Sci 2023; 24:11956. [PMID: 37569330 PMCID: PMC10418752 DOI: 10.3390/ijms241511956] [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: 06/12/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 08/13/2023] Open
Abstract
Cells produce free radicals and antioxidants when exposed to toxic compounds during cellular metabolism. However, free radicals are deleterious to lipids, proteins, and nucleic acids. Antioxidants neutralize and eliminate free radicals from cells, preventing cell damage. Therefore, the study aims to determine whether the antioxidants butylated hydroxyanisole (BHA) and butylated hydroxytoluene (BHT) will ameliorate the maximum dose of acrylamide and alpha (α)-solanine synergistic toxic effects in exposed BEAS-2B cells. These toxic compounds are consumed worldwide by eating potato products. BEAS-2B cells were simultaneously treated with BHA 10 μM and BHT 20 μM and incubated in a 5% CO2 humidified incubator for 24 h, followed by individual or combined treatment with acrylamide (3.5 mM) and α-solanine (44 mM) for 48 h, including the controls. Cell morphology, DNA, RNA, and protein were analyzed. The antioxidants did not prevent acrylamide and α-solanine synergistic effects in exposed BEAS-2B cells. However, cell morphology was altered; polymerase chain reaction (PCR) showed reduced RNA constituents but not DNA. In addition, the toxic compounds synergistically inhibited AKT/PKB expression and its downstream genes. The study showed BHA and BHT are not protective against the synergetic toxic effects of acrylamide and α-solanine in exposed BEAS-2B cells.
Collapse
Affiliation(s)
- Hoda Awad Eltayeb
- Department of Biology, Texas Southern University, Houston, TX 77004, USA
- Department of Environmental and Interdisciplinary Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Leandra Stewart
- Department of Biology, Texas Southern University, Houston, TX 77004, USA
| | - Mounira Morgem
- Department of Biology, Texas Southern University, Houston, TX 77004, USA
- Department of Environmental and Interdisciplinary Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Tommie Johnson
- Department of Biology, Texas Southern University, Houston, TX 77004, USA
- Department of Environmental and Interdisciplinary Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Michael Nguyen
- Department of Biology, Texas Southern University, Houston, TX 77004, USA
| | - Kadeshia Earl
- Department of Biology, Texas Southern University, Houston, TX 77004, USA
- Department of Environmental and Interdisciplinary Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Ayodotun Sodipe
- Department of Biology, Texas Southern University, Houston, TX 77004, USA
| | - Desirée Jackson
- Department of Biology, Texas Southern University, Houston, TX 77004, USA
| | | |
Collapse
|
7
|
Determination of trace metal ions in Gentiana rigescens by inductively coupled plasma-optical emission spectrometry after deep eutectic solvent-based digestion and related pharmacodynamic evaluation. Anal Chim Acta 2022; 1221:340109. [DOI: 10.1016/j.aca.2022.340109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/14/2022] [Accepted: 06/19/2022] [Indexed: 11/22/2022]
|
8
|
Giacalone AG, Merritt ME, Ragavan M. Ex Vivo Hepatic Perfusion Through the Portal Vein in Mouse. J Vis Exp 2022:10.3791/63154. [PMID: 35343950 PMCID: PMC9258001 DOI: 10.3791/63154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Metabolic diseases such as diabetes, pre-diabetes, non-alcoholic fatty liver disease (NAFLD), and nonalcoholic steatohepatitis (NASH) are becoming increasingly common. Ex vivo liver perfusions allow for a comprehensive analysis of liver metabolism using nuclear magnetic resonance (NMR), in nutritional conditions that can be rigorously controlled. As in silico simulations remain a primarily theoretical means of assessing hormone actions and the effects of pharmaceutical intervention, the perfused liver remains one of the most valuable test beds for understanding hepatic metabolism. As these studies guide basic insights into hepatic physiology, results must be accurate and reproducible. The greatest factor in the reproducibility of ex vivo hepatic perfusion is the quality of surgery. Therefore, we have introduced an organized and streamlined method to perform ex vivo mouse liver perfusions in the context of in situ NMR experiments. We also describe a unique application and discuss common issues encountered in these studies. The overall purpose is to provide an uncomplicated guide to a technique we have refined over several years that we deem the golden standard for obtaining reproducible results in hepatic resections and perfusions in the context of in situ NMR experiments. The distance to the center of the field for the magnet as well as the inaccessibility of the tissue to intervention during the NMR experiment makes our methods novel.
Collapse
Affiliation(s)
| | - Matthew E Merritt
- Department of Biochemistry and Molecular Biology, University of Florida
| | - Mukundan Ragavan
- Department of Biochemistry and Molecular Biology, University of Florida;
| |
Collapse
|
9
|
Gu F, Krüger A, Roggenkamp HG, Alpers R, Lodygin D, Jaquet V, Möckl F, Hernandez C LC, Winterberg K, Bauche A, Rosche A, Grasberger H, Kao JY, Schetelig D, Werner R, Schröder K, Carty M, Bowie AG, Huber S, Meier C, Mittrücker HW, Heeren J, Krause KH, Flügel A, Diercks BP, Guse AH. Dual NADPH oxidases DUOX1 and DUOX2 synthesize NAADP and are necessary for Ca 2+ signaling during T cell activation. Sci Signal 2021; 14:eabe3800. [PMID: 34784249 DOI: 10.1126/scisignal.abe3800] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The formation of Ca2+ microdomains during T cell activation is initiated by the production of nicotinic acid adenine dinucleotide phosphate (NAADP) from its reduced form NAADPH. The reverse reaction—NAADP to NAADPH—is catalyzed by glucose 6-phosphate dehydrogenase (G6PD). Here, we identified NADPH oxidases NOX and DUOX as NAADP-forming enzymes that convert NAADPH to NAADP under physiological conditions in vitro. T cells express NOX1, NOX2, and, to a minor extent, DUOX1 and DUOX2. Local and global Ca2+ signaling were decreased in mouse T cells with double knockout of Duoxa1 and Duoxa2 but not with knockout of Nox1 or Nox2. Ca2+ microdomains in the first 15 s upon T cell activation were significantly decreased in Duox2−/− but not in Duox1−/− T cells, whereas both DUOX1 and DUOX2 were required for global Ca2+ signaling between 4 and 12 min after stimulation. Our findings suggest that a DUOX2- and G6PD-catalyzed redox cycle rapidly produces and degrades NAADP through NAADPH as an inactive intermediate.
Collapse
Affiliation(s)
- Feng Gu
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Aileen Krüger
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hannes G Roggenkamp
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Rick Alpers
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Dmitri Lodygin
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Vincent Jaquet
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Franziska Möckl
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Lola C Hernandez C
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Kai Winterberg
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andreas Bauche
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anette Rosche
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Helmut Grasberger
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| | - John Y Kao
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel Schetelig
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - René Werner
- Department of Computational Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Katrin Schröder
- Institute of Cardiovascular Physiology, Goethe-Universität, 60590 Frankfurt, Germany
| | - Michael Carty
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Samuel Huber
- Department of Gastroenterology with Sections Infectiology and Tropical Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Chris Meier
- Organic Chemistry, University of Hamburg, 20146 Hamburg, Germany
| | - Hans-Willi Mittrücker
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Joerg Heeren
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Karl-Heinz Krause
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Björn-Philipp Diercks
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Andreas H Guse
- Calcium Signaling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
10
|
Huang Y, Deng S, Ai Y, Mo Y, Li W, Peng Q, Huang L, Zhang L. MicroRNA-125b alleviates hydrogen-peroxide-induced abnormal mitochondrial dynamics in HT22 cells by inhibiting p53. Metab Brain Dis 2021; 36:601-608. [PMID: 33475982 DOI: 10.1007/s11011-020-00666-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 12/29/2020] [Indexed: 11/30/2022]
Abstract
Micro-RNA125b (miR-125b) and tumor protein p53 (p53) are involved in the regulation of mitochondrial dynamics; however, the mechanism of their possible interaction during oxidative stress remains unclear. In this study, we investigated the role and mechanism of miR-125b and p53 in oxidative stress-induced mitochondrial damage in immortalized mouse hippocampal HT22 cells. Following stimulation with H2O2, we observed downregulation of miR-125b expression, upregulation of p53 expression, mitochondria were damaged and increased cell death. Overexpression of miR-125b alleviated mitochondrial damage and inhibited p53 expression. Furthermore, confocal and electron microscopy showed that overexpression of p53 eliminated the protective effect of miR-125b on the mitochondria. Thus, miR-125b alleviates abnormal mitochondrial homeostasis in H2O2-treated HT22 cells by suppressing p53 expression. Our data reveal a new model by which miR-125b influences mitochondrial dynamics.
Collapse
Affiliation(s)
- Yan Huang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008
| | - Songyun Deng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008
| | - Yuhang Ai
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008
| | - Yunan Mo
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008
| | - Wenchao Li
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008
| | - Qianyi Peng
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008
| | - Li Huang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008
| | - Lina Zhang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China, 410008.
| |
Collapse
|
11
|
Fang Y, Xing C, Wang X, Cao H, Zhang C, Guo X, Zhuang Y, Hu R, Hu G, Yang F. Activation of the ROS/HO-1/NQO1 signaling pathway contributes to the copper-induced oxidative stress and autophagy in duck renal tubular epithelial cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 757:143753. [PMID: 33316526 DOI: 10.1016/j.scitotenv.2020.143753] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/28/2020] [Accepted: 10/31/2020] [Indexed: 06/12/2023]
Abstract
The aim of this study was to investigate the crosstalk between oxidative stress and autophagy through the ROS/HO-1/NQO1 pathway caused by copper (Cu). Duck renal tubular epithelial cells were treated in Cu sulfate (CuSO4) (0, 100 and 200 μM) for 12 h, and in the combination of CuSO4 (200 μM) and reactive oxygen species (ROS) scavenger (butyl hydroxyanisole, BHA, 100 μM), or HO-1 inhibitor (zinc protoporphyrin, ZnPP, 10 μM) for 12 h. Results revealed that Cu could significantly elevate the levels of intracellular ROS, superoxide dismutase, hydrogen peroxide, malondialdehyde, glutathione, simultaneously reduce catalase and glutathione peroxidase levels, and upregulate HO-1, SOD-1, CAT, NQO1, GCLM mRNA levels and HO-1, SOD-1 protein levels. Additionally, Cu could observably increase the number of autophagosomes, acidic vesicle organelles (AVOs) and LC3 puncta; upregulate mRNA levels of mTOR, Beclin-1, ATG7, ATG5, ATG3, LC3II and protein levels of Beclin-1, LC3II/LC3I, downregulate LC3I mRNA level. Both treatments with BHA and ZnPP could significantly alleviate the changes of antioxidant indexes levels and ROS accumulation, reduce the increase of the number of autophagosomes, AVOs and LC3 puncta, and mitigate the above changed oxidative stress and autophagy related mRNA and protein levels induced by Cu. In summary, our findings indicated that excessive Cu could induce oxidative stress and autophagy by activating the ROS/HO-1/NQO1 pathway, and inhibition of HO-1 might attenuate Cu-induced oxidative stress and autophagy in duck renal tubular epithelial cells.
Collapse
Affiliation(s)
- Yukun Fang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Chenghong Xing
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Xiaoyu Wang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Huabin Cao
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Xiaoquan Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Yu Zhuang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - RuiMing Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Guoliang Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China
| | - Fan Yang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, No. 1101 Zhimin Avenue, Economic and Technological Development District, Nanchang 330045, Jiangxi, PR China.
| |
Collapse
|
12
|
Lion's Mane Mushroom, Hericium erinaceus (Bull.: Fr.) Pers. Suppresses H 2O 2-Induced Oxidative Damage and LPS-Induced Inflammation in HT22 Hippocampal Neurons and BV2 Microglia. Antioxidants (Basel) 2019; 8:antiox8080261. [PMID: 31374912 PMCID: PMC6720269 DOI: 10.3390/antiox8080261] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/28/2019] [Accepted: 08/01/2019] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress and inflammation in neuron-glia system are key factors in the pathogenesis of neurodegenerative diseases. As synthetic drugs may cause side effects, natural products have gained recognition for the prevention or management of diseases. In this study, hot water (HE-HWA) and ethanolic (HE-ETH) extracts of the basidiocarps of Hericium erinaceus mushroom were investigated for their neuroprotective and anti-inflammatory activities against hydrogen peroxide (H2O2)-induced neurotoxicity in HT22 mouse hippocampal neurons and lipopolysaccharide (LPS)-induced BV2 microglial activation respectively. HE-ETH showed potent neuroprotective activity by significantly (p < 0.0001) increasing the viability of H2O2-treated neurons. This was accompanied by significant reduction in reactive oxygen species (ROS) (p < 0.05) and improvement of the antioxidant enzyme catalase (CAT) (p < 0.05) and glutathione (GSH) content (p < 0.01). Besides, HE-ETH significantly improved mitochondrial membrane potential (MMP) (p < 0.05) and ATP production (p < 0.0001) while reducing mitochondrial toxicity (p < 0.001), Bcl-2-associated X (Bax) gene expression (p < 0.05) and nuclear apoptosis (p < 0.0001). However, gene expression of Nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase 1 (HO-1) and NAD(P)H quinone dehydrogenase 1 (NQO1) were unaffected (p > 0.05). HE-ETH also significantly (p < 0.0001) reduced nitric oxide (NO) level in LPS-treated BV2 indicating an anti-inflammatory activity in the microglia. These findings demonstrated HE-ETH maybe a potential neuroprotective and anti-inflammatory agent in neuron-glia environment.
Collapse
|
13
|
Ding H, Wen Z. Overexpression of C‑sis inhibits H2O2‑induced Buffalo rat liver cell apoptosis in vitro and alleviates liver injury in a rat model of fulminant hepatic failure. Int J Mol Med 2018; 42:873-882. [PMID: 29786113 PMCID: PMC6034937 DOI: 10.3892/ijmm.2018.3684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 05/04/2018] [Indexed: 01/17/2023] Open
Abstract
The present study aimed to investigate the role of the C‑sis gene in the apoptosis of hepatocytes in vitro and in the liver function of a rat model of fulminant hepatic failure (FHF). Buffalo rat liver (BRL) cells were treated with hydrogen peroxide (H2O2) to induce apoptosis and then transfected with a C‑sis overexpression vector. A rat model of FHF was established, and C‑sis was overexpressed. The mRNA and protein expression of C‑sis were examined using reverse transcription‑polymerase chain reaction and western blot analyses, respectively. Cell viability was assessed by CCK8, and a TUNEL assay was used to examine cell apoptosis. Flow cytometry was used for cell cycle detection. Hematoxylin and eosin staining was used for histological examination. The levels of alanine transaminase (ALT) and aspartate transaminase (AST) were also examined in the rats. The results showed that C‑sis was successfully overexpressed in the cells and rat model. Compared with H2O2‑treated BRL cells, the overexpression of C‑sis significantly inhibited cell apoptosis, promoted cell viability, and decreased the expression of cleaved caspase-3. Similar results were observed in the FHF rats treated with the C‑sis overexpression plasmid, compared with those treated with empty plasmids. In addition, in the FHF rats overexpressing C‑sis, histological examination showed that liver injury was alleviated, the levels of ALT and AST were significantly decreased, and mortality rate was significantly decreased, compared with those observed in the rats treated with empty plasmids. In conclusion, the overexpression of C‑sis inhibited the H2O2‑induced apoptosis of BRL cells in vitro, and alleviated liver injury, improved liver function, and decreased mortality rates in rat models of FHF.
Collapse
Affiliation(s)
- Hao Ding
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhili Wen
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
14
|
Santos Felix AC, Novaes CG, Pires Rocha M, Barreto GE, do Nascimento BB, Giraldez Alvarez LD. Mixture Design and Doehlert Matrix for the Optimization of the Extraction of Phenolic Compounds from Spondias mombin L Apple Bagasse Agroindustrial Residues. Front Chem 2018; 5:116. [PMID: 29354632 PMCID: PMC5760567 DOI: 10.3389/fchem.2017.00116] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/29/2017] [Indexed: 01/12/2023] Open
Abstract
In this study, we have determined, using RSM (mixture design and Doehlert matrix), the optimum values of the independent variables to achieve the maximum response for the extraction of total phenolic compounds from Spondias mombin L bagasse agroindustrial residues in order to preserve their antioxidant activity. The extraction of phenolic compounds, as well as their antioxidant capacity and the capacity to scavenge ABTS, was determined by the modified DPPH method at different periods of time, temperature, velocity of rotation and solvents concentration. We observed that the optimum condition for the highest antioxidant yield was obtained using water (60.84%), acetone (30.31%), and ethanol (8.85%) at 30°C during 20 min at 50 rpm. We have also found that the maximum yield of total phenolics was 355.63 ± 9.77 (mg GAE/100 g), showing an EC50 of 3,962.24 ± 41.20 (g fruit/g of DPPH) and 8.36 ± 0.30 (μM trolox/g fruit), which were measured using DPPH and ABTS assays. These results suggest that RSM was successfully applied for optimizing the extraction of phenolics compounds thus preserving their antioxidant activity.
Collapse
Affiliation(s)
- Antonio C Santos Felix
- Grupo de Pesquisa Aromas e Análise de Alimentos, Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Jequié, Brazil
| | - Cleber G Novaes
- Grupo de Pesquisa Laboratório de Química Analítica, Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Jequié, Brazil
| | - Maísla Pires Rocha
- Grupo de Pesquisa Aromas e Análise de Alimentos, Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Jequié, Brazil
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia.,Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Baraquizio B do Nascimento
- Grupo de Pesquisa Aromas e Análise de Alimentos, Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Jequié, Brazil
| | - Lisandro D Giraldez Alvarez
- Grupo de Pesquisa Aromas e Análise de Alimentos, Departamento de Ciências e Tecnologias, Universidade Estadual do Sudoeste da Bahia, Jequié, Brazil
| |
Collapse
|
15
|
Elmansi A, El-Karef A, Shishtawy M, Eissa L. Hepatoprotective Effect of Curcumin on Hepatocellular Carcinoma Through Autophagic and Apoptic Pathways. Ann Hepatol 2017; 16:607-618. [PMID: 28611265 DOI: 10.5604/01.3001.0010.0307] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND RATIONALE Microtubule-associated protein light chain 3-II (LC3-II), and Sequestosome-1 (SQSTM1) are proteins that can be used as markers for autophagic pathway. Bcl-2 protein is reported to be inversely correlated with apoptosis. We aimed to investigate the effects of curcumin on liver inflammation and fibrosis up to the first dysplastic stage of Hepatocellular carcinoma (HCC) induced by Thioacetamide (TAA) in rats and to clarify the effects of curcumin on LC3-II, SQSTM1, and Bcl-2. Male Sprague-Dawley rats were randomized into four groups: Control group, TAA group, Curcumin low-dose group, and Curcumin highdose group. The last three groups received TAA 200 mg/kg i.p. twice weekly for 18 weeks. Oxidative stress markers as hepatic malondialdehyde (MDA) concentration and superoxide dismutase (SOD) activity were measured by colorimetric methods. Hepatic SQSTM1 concentration was measured by ELISA, and gene expression levels of Bcl-2, and LC3-II were measured by RT-PCR.We also investigated the in vitro effect of curcumin on HepG2 cells viability through MTT assay, and the involvement of autophagy in this effect. RESULTS Curcumin increased the survival percent in rats, decreased -fetoprotein (AFP) concentration, and serum aspartate aminotransferase (AST) activity, and increased serum albumin concentration. Curcumin also significantly reduced oxidative stress in liver, inhibited apoptosis, and induced autophagy. In vitro, curcumin (50 µM) decreased HepG2 cells viabilityand the concentration of SQSTM1. CONCLUSIONS Curcumin leads to protection against TAA induced HCC up to the first dysplastic stage through activating autophagic pathway and inhibiting apoptosis. Also, the antioxidant activity of curcumin almost prevents liver fibrosis.
Collapse
MESH Headings
- Animals
- Anticarcinogenic Agents/pharmacology
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis/drug effects
- Autophagy/drug effects
- Carcinoma, Hepatocellular/chemically induced
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/prevention & control
- Cell Survival/drug effects
- Cell Transformation, Neoplastic/chemically induced
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Curcumin/pharmacology
- Dose-Response Relationship, Drug
- Hep G2 Cells
- Humans
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Liver Neoplasms/chemically induced
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/prevention & control
- Male
- Microtubule-Associated Proteins/metabolism
- Oxidative Stress/drug effects
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Rats, Sprague-Dawley
- Sequestosome-1 Protein/metabolism
- Signal Transduction/drug effects
- Thioacetamide
- Time Factors
Collapse
Affiliation(s)
- Ahmed Elmansi
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Amro El-Karef
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mamdouh Shishtawy
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Laila Eissa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
16
|
Promotion of behavior and neuronal function by reactive oxygen species in C. elegans. Nat Commun 2016; 7:13234. [PMID: 27824033 PMCID: PMC5105148 DOI: 10.1038/ncomms13234] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/14/2016] [Indexed: 12/21/2022] Open
Abstract
Reactive oxygen species (ROS) are well known to elicit a plethora of detrimental effects on cellular functions by causing damages to proteins, lipids and nucleic acids. Neurons are particularly vulnerable to ROS, and nearly all forms of neurodegenerative diseases are associated with oxidative stress. Here, we report the surprising finding that exposing C. elegans to low doses of H2O2 promotes, rather than compromises, sensory behavior and the function of sensory neurons such as ASH. This beneficial effect of H2O2 is mediated by an evolutionarily conserved peroxiredoxin-p38/MAPK signaling cascade. We further show that p38/MAPK signals to AKT and the TRPV channel OSM-9, a sensory channel in ASH neurons. AKT phosphorylates OSM-9, and such phosphorylation is required for H2O2-induced potentiation of sensory behavior and ASH neuron function. Our results uncover a beneficial effect of ROS on neurons, revealing unexpected complexity of the action of oxidative stressors in the nervous system. The deleterious role of reactive oxygen species has been widely reported in the nervous system. Here the authors report that surprisingly, low doses of H2O2 in fact enhances sensory neuron function and promotes sensory behaviors in C. elegans.
Collapse
|
17
|
Li H, Zhou X, Wu M, Deng M, Wang C, Hou J, Mou P. The cytotoxicity and protective effects of Astragalus membranaceus extracts and butylated hydroxyanisole on hydroxyl radical-induced apoptosis in fish erythrocytes. ACTA ACUST UNITED AC 2016; 2:376-382. [PMID: 29767041 PMCID: PMC5941053 DOI: 10.1016/j.aninu.2016.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 08/08/2016] [Indexed: 12/22/2022]
Abstract
Erythrocytes play an essential role in transporting O2 and CO2 for respiration in fish. However, erythrocytes continuously suffer from reactive oxygen species (ROS) -induced oxidative stress and apoptosis. Thus, it is essential to expand our knowledge of how to protect erythrocytes against ROS-induced oxidative stress and apoptosis in fish. In this study, we explored the cytotoxicity and the effects of butylated hydroxyanisole (BHA), ethyl ether extracts, ethyl acetate extracts, acetone extracts (AE), ethanol extracts, and aqueous extracts of Astragalus membranaceus (EAm) on hydroxyl radical (•OH)-induced apoptosis in carp erythrocytes. The rat hepatocytes and carp erythrocytes were incubated with different concentrations of BHA or EAm(0.125 to 1 mg/mL). The toxicity in rat hepatocytes and carp erythrocytes was then measured using a 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay and a haemolysis assay, respectively. The carp erythrocytes were treated with BHA or EAm in the presence of 40 μmol/L FeSO4 and 20 μmol/L H2O2 at 37 °C, except for the control group. Oxidative stress and apoptosis parameters in the carp erythrocytes were then evaluated using the commercial kit. The results indicated that at high concentrations, BHA and EAm could induce toxicity in rat hepatocytes and fish erythrocytes. However, BHA was more toxic than EAm at the same concentrations. Moreover, the toxicity order of BHA and EAm in the fish erythrocytes approximately agreed with that for the rat hepatocytes. Butylated hydroxyanisole and EAm suppressed the •OH-induced phosphatidylserine exposure and DNA fragmentation (the biomarkers of apoptosis) by decreasing the generation of ROS, inhibiting the oxidation of cellular components, and restoring the activities of antioxidants in carp erythrocytes. Of all of the examined EAm, the AE showed the strongest effects. The effects of AE on superoxide anion, H2O2, met-haemoglobin and reduced glutathione levels, as well as glutathione reductase activity and apoptosis were equivalent to or stronger than those of BHA. These results revealed that the AE of Astragalus membranaceus could be used as a potential natural antioxidant or apoptosis inhibitor in fish erythrocytes.
Collapse
Affiliation(s)
- Huatao Li
- College of Life Sciences, Neijiang Normal University, Neijiang 641000, China.,Conservation and Utilization of Fishes Resources in the Upper Reaches of the Yangtze River Key Laboratory of Sichuan Province, Neijiang Normal University, Neijiang 641000, China
| | - Xiaoqiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Min Wu
- Archives, Neijiang Normal University, Neijiang 641000, China
| | - Mengling Deng
- College of Life Sciences, Neijiang Normal University, Neijiang 641000, China.,Conservation and Utilization of Fishes Resources in the Upper Reaches of the Yangtze River Key Laboratory of Sichuan Province, Neijiang Normal University, Neijiang 641000, China
| | - Chao Wang
- College of Life Sciences, Neijiang Normal University, Neijiang 641000, China.,Conservation and Utilization of Fishes Resources in the Upper Reaches of the Yangtze River Key Laboratory of Sichuan Province, Neijiang Normal University, Neijiang 641000, China
| | - Jingjing Hou
- College of Life Sciences, Neijiang Normal University, Neijiang 641000, China.,Conservation and Utilization of Fishes Resources in the Upper Reaches of the Yangtze River Key Laboratory of Sichuan Province, Neijiang Normal University, Neijiang 641000, China
| | - Pengju Mou
- College of Life Sciences, Neijiang Normal University, Neijiang 641000, China.,Conservation and Utilization of Fishes Resources in the Upper Reaches of the Yangtze River Key Laboratory of Sichuan Province, Neijiang Normal University, Neijiang 641000, China
| |
Collapse
|