1
|
Glorieux C, Buc Calderon P. Targeting catalase in cancer. Redox Biol 2024; 77:103404. [PMID: 39447253 PMCID: PMC11539659 DOI: 10.1016/j.redox.2024.103404] [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: 08/26/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Healthy cells have developed a sophisticated network of antioxidant molecules to prevent the toxic accumulation of reactive oxygen species (ROS) generated by diverse environmental stresses. On the opposite, cancer cells often exhibit high levels of ROS and an altered levels of antioxidant molecules compared to normal cells. Among them, the antioxidant enzyme catalase plays an essential role in cell defense against oxidative stress through the dismutation of hydrogen peroxide into water and molecular oxygen, and its expression is often decreased in cancer cells. The elevation of ROS in cancer cells provides them proliferative advantages, and leads to metabolic reprogramming, immune escape and metastasis. In this context, catalase is of critical importance to control these cellular processes in cancer through various mechanisms. In this review, we will discuss the major progresses and challenges in understanding the role of catalase in cancer for this last decade. This review also aims to provide important updates regarding the regulation of catalase expression, subcellular localization and discuss about the potential role of microbial catalases in tumor environment. Finally, we will describe the different catalase-based therapies and address the advantages, disadvantages, and limitations associated with modulating catalase therapeutically in cancer treatment.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060, Guangzhou, China.
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de La Salud, Universidad Arturo Prat, 1100000, Iquique, Chile; Instituto de Química Medicinal, Universidad Arturo Prat, 1100000, Iquique, Chile; Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université Catholique de Louvain, 1200, Brussels, Belgium.
| |
Collapse
|
2
|
Gerges MN, Abdou FY, El Gamal DM, Habieb ME. Metformin ameliorates vanadium pentoxide or gamma irradiation-stimulated hepatotoxicity in male rats via targeting endoplasmic reticulum stress-induced apoptosis. Hum Exp Toxicol 2024; 43:9603271241307859. [PMID: 39648841 DOI: 10.1177/09603271241307859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
INTRODUCTION This work aims to validate the ameliorative influence of metformin against endoplasmic reticulum stress (ERS)-prompted apoptosis caused by vanadium pentoxide (V2O5) or gamma-irradiation (γ-irradiation) in hepatic tissues of male rats. METHODS There were six groups of rats: the control, metformin (100 mg/kg body weight, i.p.), V2O5 (12.5 mg/kg body weight, i.p), V2O5 plus metformin, γ-irradiation group (acute dose 6 Gy), and γ-irradiation plus metformin; for 2 weeks. Hepatic malondialdehyde (MDA) and reduced glutathione (GSH) levels were evaluated. Additionally, the protein expression of certain endoplasmic reticulum stress-related (ERS) biomarkers; Inositol requirement enzyme 1α (IRE1α), TNF receptor-associated factor 2 (TRAF2), and Apoptosis signal-regulating kinase 1 (ASK1); were estimated in hepatic tissues. Moreover, apoptosis-associated biomarkers; Bax, Bcl-2, caspase-3 and HSP70 levels have been assessed. Furthermore, histopathological changes in hepatic tissues were observed. RESULTS Metformin with V2O5 or γ-irradiation significantly decreased MDA, IRE1α, TRAF2, ASK1, Bax, and caspase-3 compared with V2O5 or γ-irradiated groups. Meanwhile, it significantly elevated GSH, Bcl-2, and HSP70 levels compared to exposure to V2O5 or γ-irradiation groups. Interestingly, the obtained results concur well with histological alterations. DISCUSSION Our findings demonstrate the protective influence of metformin against ER stress-induced apoptosis through enhancing GSH and reduction of ERS and apoptosis suggesting that metformin may have positive impacts as a potential radiation protector beyond its glucose-lowering effect.
Collapse
Affiliation(s)
- Marian N Gerges
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Fatma Y Abdou
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| | - Doaa M El Gamal
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mahmoud E Habieb
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, Egypt
| |
Collapse
|
3
|
Inceu AI, Neag MA, Catinean A, Bocsan CI, Craciun CI, Melincovici CS, Muntean DM, Onofrei MM, Pop RM, Buzoianu AD. The Effects of Probiotic Bacillus Spores on Dexamethasone-Treated Rats. Int J Mol Sci 2023; 24:15111. [PMID: 37894792 PMCID: PMC10606902 DOI: 10.3390/ijms242015111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/03/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
Glucocorticoids are effective anti-inflammatory and immunosuppressive agents. Long-term exposure is associated with multiple metabolic side effects. Spore-forming probiotic bacteria have shown modulatory properties regarding glycolipid metabolism and inflammation. The aim of this study was to evaluate, for the first time, the effects of Bacillus species spores (B. licheniformis, B. indicus, B. subtilis, B. clausii, and B. coagulans) alone and in combination with metformin against dexamethasone-induced systemic disturbances. A total of 30 rats were randomly divided into 5 groups: group 1 served as control (CONTROL), group 2 received dexamethasone (DEXA), group 3 received DEXA and MegaSporeBiotic (MSB), group 4 received DEXA and metformin (MET), and group 5 received DEXA, MSB, and MET. On the last day of the experiment, blood samples and liver tissue samples for histopathological examination were collected. We determined serum glucose, total cholesterol, triglycerides, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-10 (IL-10), catalase, total antioxidant capacity (TAC), and metformin concentration. DEXA administration caused hyperglycemia and hyperlipidemia, increased inflammation cytokines, and decreased antioxidant markers. Treatment with MSB reduced total cholesterol, suggesting that the administration of Bacillus spores-based probiotics to DEXA-treated rats could ameliorate metabolic parameters.
Collapse
Affiliation(s)
- Andreea Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Maria Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Adrian Catinean
- Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| | - Corina Ioana Bocsan
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Cristian Ioan Craciun
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Carmen Stanca Melincovici
- Department of Histology, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (C.S.M.); (M.M.O.)
| | - Dana Maria Muntean
- Department of Pharmaceutical Technology and Biopharmaceutics, Iuliu Hatieganu University of Medicine and Pharmacy, 400010 Cluj-Napoca, Romania;
| | - Mădălin Mihai Onofrei
- Department of Histology, Iuliu Hatieganu University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania; (C.S.M.); (M.M.O.)
| | - Raluca Maria Pop
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania; (A.I.I.); (C.I.B.); (C.I.C.); (R.M.P.); (A.D.B.)
| |
Collapse
|
4
|
Dagsuyu E, Koroglu P, Gul IB, Bulan OK, Yanardag R. Oxidative brain and cerebellum injury in diabetes and prostate cancer model: Protective effect of metformin. J Biochem Mol Toxicol 2023; 37:e23440. [PMID: 37354076 DOI: 10.1002/jbt.23440] [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: 01/25/2023] [Revised: 05/24/2023] [Accepted: 06/14/2023] [Indexed: 06/26/2023]
Abstract
The body can host the spread of prostate cancer cells. Metastases from prostate cancer are more frequently seen in the brain, liver, lungs, and lymph nodes. A well-known antidiabetic drug, metformin, is also known to have antitumor effects. Our study focuses on the evaluation of potential metformin protective effects on brain and cerebellum damage in streptozotocin (STZ)-induced diabetic and Dunning prostate cancer models. In this investigation, six groups of male Copenhagen rats were created: control, diabetic (D), cancer (C), diabetic + cancer (DC), cancer + metformin, and diabetic + cancer + metformin. The brain and cerebellum tissues of the rats were taken after sacrifice. Oxidative stress markers including reduced glutathione level, lipid peroxidation, glutathione reductase, glutathione peroxidase, glutathione-S-transferase, catalase, superoxide dismutase activities, reactive oxygen species, total oxidant and total antioxidant status, lactate dehydrogenase, xanthine oxidase, acetylcholinesterase activities, protein carbonyl contents, nitric oxide and OH-proline levels, sodium potassium ATPase, carbonic anhydrase, and glucose-6-phosphate dehydrogenase activities; glycoprotein levels including hexose, hexosamine, fucose, and sialic acid levels; and histone deacetylase activity as a cancer marker were determined. Oxidative stress markers were impaired and glycoprotein levels and histone deacetylase activity were increased in the D, C, and DC groups. Metformin therapy reversed these effects. Metformin was found to protect the brain and cerebellum of STZ-induced diabetic rats with Dunning prostate cancer from harm caused by MAT-Lylu metastatic cells.
Collapse
Affiliation(s)
- Eda Dagsuyu
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Pınar Koroglu
- Department of Histology and Embryology, Faculty of Medicine, Halic University, Istanbul, Turkey
| | - Ilknur B Gul
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Omur K Bulan
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Refiye Yanardag
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
5
|
Shi X, Li L, Liu Z, Wang F, Huang H. Exploring the mechanism of metformin action in Alzheimer's disease and type 2 diabetes based on network pharmacology, molecular docking, and molecular dynamic simulation. Ther Adv Endocrinol Metab 2023; 14:20420188231187493. [PMID: 37780174 PMCID: PMC10540612 DOI: 10.1177/20420188231187493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/19/2023] [Indexed: 10/03/2023] Open
Abstract
Background Metformin, which has been shown to be highly effective in treating type 2 diabetes (T2D), is also believed to be valuable for Alzheimer's disease (AD). Computer simulation techniques have emerged as an innovative approach to explore mechanisms. Objective To study the potential mechanism of metformin action in AD and T2D. Methods The chemical structure of metformin was obtained from PubChem. The targets of metformin were obtained from PubChem, Pharm Mapper, Batman, SwissTargetPrediction, DrugBank, and PubMed. The pathogenic genes of AD and T2D were retrieved from the GeneCards, OMIM, TTD, Drugbank, PharmGKB, and DisGeNET. The intersection of metformin with the targets of AD and T2D is represented by a Venn diagram. The protein-protein interaction (PPI) and core targets networks of intersected targets were constructed by Cytoscape 3.7.1. The enrichment information of GO and Kyoto Encyclopedia of Gene and Genomics (KEGG) pathways obtained by the Metascape was made into a bar chart and a bubble diagram. AutoDockTools, Pymol, and Chem3D were used for the molecular docking. Gromacs software was used to perform molecular dynamics (MD) simulation of the best binding target protein. Results A total of 115 key targets of metformin for AD and T2D were obtained. GO analysis showed that biological process mainly involved response to hormones and the regulation of ion transport. Cellular component was enriched in the cell body and axon. Molecular function mainly involved kinase binding and signal receptor regulator activity. The KEGG pathway was mainly enriched in pathways of cancer, neurodegeneration, and endocrine resistance. Core targets mainly included TP53, TNF, VEGFA, HIF1A, IL1B, IGF1, ESR1, SIRT1, CAT, and CXCL8. The molecular docking results showed best binding of metformin to CAT. MD simulation further indicated that the CAT-metformin complex could bind well and converge relatively stable at 30 ns. Conclusion Metformin exerts its effects on regulating oxidative stress, gluconeogenesis and inflammation, which may be the mechanism of action of metformin to improve the common pathological features of T2D and AD.
Collapse
Affiliation(s)
- Xin Shi
- Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Lingling Li
- Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Zhiyao Liu
- Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Fangqi Wang
- Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Hailiang Huang
- Shandong University of Traditional Chinese Medicine, 4655 Guyunhu Street, Changqing District, Jinan City, Shandong Province, China
| |
Collapse
|
6
|
Ayhan S, Hancerliogullari N, Guney G, Gozukucuk M, Caydere M, Guney SS, Tokmak A, Ustun Y. Does the addition of metformin to carboplatin treatment decreases ovarian reserve damage associated with carboplatin usage? J Ovarian Res 2023; 16:184. [PMID: 37660125 PMCID: PMC10474675 DOI: 10.1186/s13048-023-01259-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/13/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND We aimed to determine whether adding metformin to carboplatin treatment would reduce the damage to ovarian reserve associated with carboplatin use. METHODS We included 35 adult female non-pregnant albino Wistar rats approximately three months old, weighing 220-310 g. The rats were divided into five groups of seven rats according to the treatment they received. Carboplatin and salin was given to Group 2, and carboplatin plus metformin was given to Group 3. Group 4 was administered only metformin. Group 5 was administered only salin. Carboplatin was given to Groups 2 and 3 as a single dose on the 15th day, while metformin was given to Groups 3 and 4 during the 28-day experiment. After oophorectomy, histopathologic analyses of primordial, primary, secondary, and tertiary Graff follicles according to the epithelial cells surrounding the oocyte and total follicular number were conducted per section. Serum Anti-Mullerian Hormone (AMH), tissue catalase, and malonyl dialdehyde levels were measured and compared within each group. RESULTS The baseline and 15th-day serum AMH values of the menstrual cycle were compared among the groups, and no statistically significant differences were observed (p > 0.05). Group 3, which was given both carboplatin and metformin, had statistically significantly higher 28th-day AMH levels than Group 2, which was given only carboplatin and saline (p < 0.001). The number of primordial follicles in Group 3 was found to be statistically significantly higher than in Group 2 (p < 0.001). Tissue catalase enzyme levels in Group 3 were statistically significantly higher than in Group 2 (p < 0.001). Tissue malondialdehyde levels in Group 2 were statistically significantly higher than tissue malondialdehyde levels in Groups 3 and 4 (p < 0.001). CONCLUSIONS Metformin may attenuate carboplatin-induced ovarian damage, possibly through its antioxidative effects.
Collapse
Affiliation(s)
- Sevgi Ayhan
- Department of Obstetrics and Gynecology, University of Health Sciences, Bilkent City Hospital, Ankara, Turkey
| | - Necati Hancerliogullari
- Department of Obstetrics and Gynecology, University of Health Sciences, Bilkent City Hospital, Ankara, Turkey
| | - Gurhan Guney
- Department of Reproductive Endocrinology and Infertility, Balikesir University School of Medicine, Cagis Campus,10145, 10145, Balikesir, Turkey.
| | - Murat Gozukucuk
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Muzaffer Caydere
- Department of Pathology, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| | - Sergul Selvi Guney
- Department of Midwifery, Faculty of Health Sciences, Balikesir University, Balikesir, Turkey
| | - Aytekin Tokmak
- Department of Obstetrics and Gynecology, University of Health Sciences, Bilkent City Hospital, Ankara, Turkey
| | - Yusuf Ustun
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
7
|
Vargas-Maya NI, Olmedo-Monfil V, Ramírez-Prado JH, Reyes-Cortés R, Padilla-Vaca F, Franco B. Catalases in the pathogenesis of Sporothrix schenckii research. PeerJ 2022; 10:e14478. [PMID: 36523453 PMCID: PMC9745942 DOI: 10.7717/peerj.14478] [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: 09/08/2022] [Accepted: 11/07/2022] [Indexed: 12/12/2022] Open
Abstract
Pathogenic fungal infection success depends on the ability to escape the immune response. Most strategies for fungal infection control are focused on the inhibition of virulence factors and increasing the effectiveness of antifungal drugs. Nevertheless, little attention has been focused on their physiological resistance to the host immune system. Hints may be found in pathogenic fungi that also inhabit the soil. In nature, the saprophyte lifestyle of fungi is also associated with predators that can induce oxidative stress upon cell damage. The natural sources of nutrients for fungi are linked to cellulose degradation, which in turn generates reactive oxygen species (ROS). Overall, the antioxidant arsenal needed to thrive both in free-living and pathogenic lifestyles in fungi is fundamental for success. In this review, we present recent findings regarding catalases and oxidative stress in fungi and how these can be in close relationship with pathogenesis. Additionally, special focus is placed on catalases of Sporothrix schenckii as a pathogenic model with a dual lifestyle. It is assumed that catalase expression is activated upon exposure to H2O2, but there are reports where this is not always the case. Additionally, it may be relevant to consider the role of catalases in S. schenckii survival in the saprophytic lifestyle and why their study can assess their involvement in the survival and therefore, in the virulence phenotype of different species of Sporothrix and when each of the three catalases are required. Also, studying antioxidant mechanisms in other isolates of pathogenic and free-living fungi may be linked to the virulence phenotype and be potential therapeutic and diagnostic targets. Thus, the rationale for this review to place focus on fungal catalases and their role in pathogenesis in addition to counteracting the effect of immune system reactive oxygen species. Fungi that thrive in soil and have mammal hosts could shed light on the importance of these enzymes in the two types of lifestyles. We look forward to encouraging more research in a myriad of areas on catalase biology with a focus on basic and applied objectives and placing these enzymes as virulence determinants.
Collapse
Affiliation(s)
| | | | | | - Ruth Reyes-Cortés
- Biology Department, Universidad de Guanajuato, Guanajuato, Guanajuato, México
| | - Felipe Padilla-Vaca
- Biology Department, Universidad de Guanajuato, Guanajuato, Guanajuato, México
| | - Bernardo Franco
- Biology Department, Universidad de Guanajuato, Guanajuato, Guanajuato, México
| |
Collapse
|
8
|
Grodner B, Napiórkowska M, Pisklak DM. Catalase Inhibition by Aminoalkanol Derivatives with Potential Anti-Cancer Activity-In Vitro and In Silico Studies Using Capillary Electrophoresis Method. Int J Mol Sci 2022; 23:7123. [PMID: 35806131 PMCID: PMC9266750 DOI: 10.3390/ijms23137123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/04/2022] Open
Abstract
In this work, the investigation of type and inhibitory strength of catalase by two pairs of aminoalkanol derivatives (1,7 diEthyl- and 1,7-diMethyl-8,9-diphenyl-4-azatricyclo (5.2.1.02.6) dec-8-ene- 3,5,10-trione) has been presented. The obtained results allowed for the determination of all kinetic parameters (Km, Vmax, slope angles of Lineweaver-Burk plots, Ki and IC50) on the basis of which it was shown that all four aminoalkanol derivatives are competitive inhibitors of catalase. However, the strength of action of each of them depends on the type of substituents present in the main structure of the molecule. Subtle differences in the potency of individual derivatives were possible to detect thanks to the developed, sensitive method of capillary electrophoresis, which allowed simultaneous monitoring of the mutual changes in the concentrations of substrates and products of the reaction catalyzed by the enzyme. Detailed values of kinetic parameters showed that all derivatives are weak inhibitors of catalase, which in this case is a big advantage because each inhibition of catalase activity is associated with a greater amount of accumulated, harmful reactive oxygen species. The results of docking studies also show the convergence of the binding energies values of individual inhibitors with all kinetic parameters of the investigated catalase inhibition and thus additionally confirm the weak inhibitory strength of all four aminoalkanol derivatives.
Collapse
Affiliation(s)
- Błażej Grodner
- Department of Biochemistry and Pharmacogenomics, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland
| | - Mariola Napiórkowska
- Department of Biochemistry, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland;
| | - Dariusz Maciej Pisklak
- Department of Physical Chemistry, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland;
| |
Collapse
|
9
|
Thomas C, Wurzer L, Malle E, Ristow M, Madreiter-Sokolowski CT. Modulation of Reactive Oxygen Species Homeostasis as a Pleiotropic Effect of Commonly Used Drugs. FRONTIERS IN AGING 2022; 3:905261. [PMID: 35821802 PMCID: PMC9261327 DOI: 10.3389/fragi.2022.905261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/18/2022] [Indexed: 01/17/2023]
Abstract
Age-associated diseases represent a growing burden for global health systems in our aging society. Consequently, we urgently need innovative strategies to counteract these pathological disturbances. Overwhelming generation of reactive oxygen species (ROS) is associated with age-related damage, leading to cellular dysfunction and, ultimately, diseases. However, low-dose ROS act as crucial signaling molecules and inducers of a vaccination-like response to boost antioxidant defense mechanisms, known as mitohormesis. Consequently, modulation of ROS homeostasis by nutrition, exercise, or pharmacological interventions is critical in aging. Numerous nutrients and approved drugs exhibit pleiotropic effects on ROS homeostasis. In the current review, we provide an overview of drugs affecting ROS generation and ROS detoxification and evaluate the potential of these effects to counteract the development and progression of age-related diseases. In case of inflammation-related dysfunctions, cardiovascular- and neurodegenerative diseases, it might be essential to strengthen antioxidant defense mechanisms in advance by low ROS level rises to boost the individual ROS defense mechanisms. In contrast, induction of overwhelming ROS production might be helpful to fight pathogens and kill cancer cells. While we outline the potential of ROS manipulation to counteract age-related dysfunction and diseases, we also raise the question about the proper intervention time and dosage.
Collapse
Affiliation(s)
- Carolin Thomas
- Laboratory of Energy Metabolism Institute of Translational Medicine Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Lia Wurzer
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ernst Malle
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Michael Ristow
- Laboratory of Energy Metabolism Institute of Translational Medicine Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | | |
Collapse
|
10
|
Metformin and alpha lipoic acid ameliorate hypothyroidism and its complications in adult male rats. J Diabetes Metab Disord 2022. [DOI: 10.1007/s40200-022-01063-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Korn D, Thieme AJ, Alves VM, Yeakey M, V V B Borba J, Capuzzi SJ, Fecho K, Bizon C, Edwards SW, Chirkova R, Colvis CM, Southall NT, Austin CP, Muratov EN, Tropsha A. Defining clinical outcome pathways. Drug Discov Today 2022; 27:1671-1678. [PMID: 35182735 DOI: 10.1016/j.drudis.2022.02.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/07/2022] [Accepted: 02/14/2022] [Indexed: 12/23/2022]
Abstract
Here, we propose a broad concept of 'Clinical Outcome Pathways' (COPs), which are defined as a series of key molecular and cellular events that underlie therapeutic effects of drug molecules. We formalize COPs as a chain of the following events: molecular initiating event (MIE) → intermediate event(s) → clinical outcome. We illustrate the concept with COP examples both for primary and alternative (i.e., drug repurposing) therapeutic applications. We also describe the elucidation of COPs for several drugs of interest using the publicly accessible Reasoning Over Biomedical Objects linked in Knowledge-Oriented Pathways (ROBOKOP) biomedical knowledge graph-mining tool. We propose that broader use of COP uncovered with the help of biomedical knowledge graph mining will likely accelerate drug discovery and repurposing efforts.
Collapse
Affiliation(s)
- Daniel Korn
- Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA; UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Andrew J Thieme
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Vinicius M Alves
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Michael Yeakey
- Massachusetts College of Pharmacy and Health Sciences, Boston, MA, USA
| | - Joyce V V B Borba
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Stephen J Capuzzi
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Karamarie Fecho
- Renaissance Computing Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Chris Bizon
- Renaissance Computing Institute, University of North Carolina, Chapel Hill, NC, USA
| | | | - Rada Chirkova
- Department of Computer Science, North Carolina State University, Raleigh, NC, USA
| | - Christine M Colvis
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Noel T Southall
- Department of Computer Science, North Carolina State University, Raleigh, NC, USA
| | - Christopher P Austin
- Department of Computer Science, North Carolina State University, Raleigh, NC, USA
| | - Eugene N Muratov
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| | - Alexander Tropsha
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Alves VM, Korn D, Pervitsky V, Thieme A, Capuzzi SJ, Baker N, Chirkova R, Ekins S, Muratov EN, Hickey A, Tropsha A. Knowledge-based approaches to drug discovery for rare diseases. Drug Discov Today 2022; 27:490-502. [PMID: 34718207 PMCID: PMC9124594 DOI: 10.1016/j.drudis.2021.10.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/13/2021] [Accepted: 10/21/2021] [Indexed: 02/03/2023]
Abstract
The conventional drug discovery pipeline has proven to be unsustainable for rare diseases. Herein, we discuss recent advances in biomedical knowledge mining applied to discovering therapeutics for rare diseases. We summarize current chemogenomics data of relevance to rare diseases and provide a perspective on the effectiveness of machine learning (ML) and biomedical knowledge graph mining in rare disease drug discovery. We illustrate the power of these methodologies using a chordoma case study. We expect that a broader application of knowledge graph mining and artificial intelligence (AI) approaches will expedite the discovery of viable drug candidates against both rare and common diseases.
Collapse
Affiliation(s)
- Vinicius M Alves
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA; UNC Catalyst for Rare Diseases, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Daniel Korn
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Vera Pervitsky
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Andrew Thieme
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Stephen J Capuzzi
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nancy Baker
- ParlezChem, 123 W Union Street, Hillsborough, NC 27278, USA
| | - Rada Chirkova
- Department of Computer Science, North Carolina State University, Raleigh, NC 27695-8206, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Eugene N Muratov
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pharmaceutical Sciences, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| | - Anthony Hickey
- UNC Catalyst for Rare Diseases, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Alexander Tropsha
- Laboratory for Molecular Modeling, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
13
|
Cheng FF, Liu YL, Du J, Lin JT. Metformin's Mechanisms in Attenuating Hallmarks of Aging and Age-Related Disease. Aging Dis 2022; 13:970-986. [PMID: 35855344 PMCID: PMC9286921 DOI: 10.14336/ad.2021.1213] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/13/2021] [Indexed: 11/01/2022] Open
Affiliation(s)
- Fang-Fang Cheng
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China.
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, China.
| | - Yan-Li Liu
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, China.
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, China.
| | - Jang Du
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, China.
| | - Jun-Tang Lin
- Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang 453003, China.
- Correspondence should be addressed to: Dr. Jun-Tang Lin, Stem Cell and Biotherapy Engineering Research Center of Henan, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
14
|
Réggami Y, Benkhaled A, Boudjelal A, Berredjem H, Amamra A, Benyettou H, Larabi N, Senator A, Siracusa L, Ruberto G. Artemisia herba-alba aqueous extract improves insulin sensitivity and hepatic steatosis in rodent model of fructose-induced metabolic syndrome. Arch Physiol Biochem 2021; 127:541-550. [PMID: 31464524 DOI: 10.1080/13813455.2019.1659825] [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] [Indexed: 12/13/2022]
Abstract
CONTEXT Fructose consumption is associated with the development of obesity and metabolic syndrome (MetS) in human and animal models. OBJECTIVE This study investigates the ability of an aqueous extract of Artemisia herba-alba Asso (AH) to ameliorate fructose-induced MetS in Male Wistar rats. METHODS AH extract at doses of 100, 200 and 400 mg/kg b.w./day was administered for six weeks to MetS animals. RESULTS Liquid fructose (10% w/v) intake did not vary total animal body weight, whereas, it produced moderate hyperglycemia associated with metabolic and histological alterations. Treating MetS rats with AH extract improved insulin sensitivity, alleviated atherogenic dyslipidaemia and decreased lipid deposition in their hepatic tissues. Additionally, AH extract was found to raise GSH level and antioxidant enzymes (GPx, GST and CAT) activities in rat livers homogenates. CONCLUSION The results here reported demonstrated, for the first time, that A. herba-alba have therapeutic proprieties against fructose-induced MetS in rodent model.
Collapse
Affiliation(s)
- Yassine Réggami
- Department of Microbiology and Biochemistry, Faculty of Sciences, Mohamed Boudiaf-M'sila University, M'sila, Algeria
- Laboratory of Biochemistry and Applied Microbiology, Department of Biochemistry, Faculty of Sciences, Badji Mokhtar-Annaba University, Annaba, Algeria
| | - Abderrahim Benkhaled
- Department of Microbiology and Biochemistry, Faculty of Sciences, Mohamed Boudiaf-M'sila University, M'sila, Algeria
| | - Amel Boudjelal
- Department of Microbiology and Biochemistry, Faculty of Sciences, Mohamed Boudiaf-M'sila University, M'sila, Algeria
| | - Hajira Berredjem
- Laboratory of Biochemistry and Applied Microbiology, Department of Biochemistry, Faculty of Sciences, Badji Mokhtar-Annaba University, Annaba, Algeria
| | - Amani Amamra
- Laboratory of Biochemistry and Applied Microbiology, Department of Biochemistry, Faculty of Sciences, Badji Mokhtar-Annaba University, Annaba, Algeria
| | - Halima Benyettou
- Department of Microbiology and Biochemistry, Faculty of Sciences, Mohamed Boudiaf-M'sila University, M'sila, Algeria
| | - Nadia Larabi
- Department of Microbiology and Biochemistry, Faculty of Sciences, Mohamed Boudiaf-M'sila University, M'sila, Algeria
| | - Abderrahmane Senator
- Laboratory of Applied Biochemistry, Faculty of Natural and Life Sciences, Ferhat Abbas-Setif University, Setif, Algeria
| | - Laura Siracusa
- Istituto di Chimica Biomolecolare del Consiglio Nazionale delle Ricerche (ICB-CNR), Catania, Italy
| | - Giuseppe Ruberto
- Istituto di Chimica Biomolecolare del Consiglio Nazionale delle Ricerche (ICB-CNR), Catania, Italy
| |
Collapse
|
15
|
Boonphang O, Ontawong A, Pasachan T, Phatsara M, Duangjai A, Amornlerdpison D, Jinakote M, Srimaroeng C. Antidiabetic and Renoprotective Effects of Coffea arabica Pulp Aqueous Extract through Preserving Organic Cation Transport System Mediated Oxidative Stress Pathway in Experimental Type 2 Diabetic Rats. Molecules 2021; 26:molecules26071907. [PMID: 33800673 PMCID: PMC8037495 DOI: 10.3390/molecules26071907] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/20/2021] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Abstract
Coffea arabica pulp (CP) is a by-product of coffee processing. CP contains polyphenols that have exhibited beneficial effects, including antioxidant and lipid-lowering effects, as well as enhanced insulin sensitivity, in in vitro and in vivo models. How polyphenols, as found in CP aqueous extract (CPE), affect type 2 diabetes (T2D) has not been investigated. Thus, the present study examined the potential antidiabetic, antioxidant, and renoprotective effects of CPE-rich polyphenols, using an experimental model of T2D in rats induced by a high-fat diet and a single low dose of streptozotocin. The T2D rats received either 1000 mg/kg body weight (BW) of CPE, 30 mg/kg BW of metformin (Met), or a combination treatment (CPE + Met) for 3 months. Plasma parameters, kidney morphology and function, and renal organic transport were determined. Significant hyperglycemia, hypertriglyceridemia, insulin resistance, increased renal lipid content and lipid peroxidation, and morphological kidney changes related to T2D were restored by both CPE and CPE + Met treatments. Additionally, the renal uptake of organic cation, 3H-1-methyl-4-phenylpyridinium (MPP+), was reduced in T2D, while transport was restored by CPE and CPE + Met, through an up-regulation of antioxidant genes and protein kinase Cα deactivation. Thus, CPE has antidiabetic and antioxidant effects that potentially ameliorate kidney function in T2D by preserving renal organic cation transport through an oxidative stress pathway.
Collapse
Affiliation(s)
- Oranit Boonphang
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (O.B.); (T.P.)
| | - Atcharaporn Ontawong
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (A.O.); (A.D.)
| | - Tipthida Pasachan
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (O.B.); (T.P.)
| | - Manussabhorn Phatsara
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Acharaporn Duangjai
- Division of Physiology, School of Medical Sciences, University of Phayao, Phayao 56000, Thailand; (A.O.); (A.D.)
| | - Doungporn Amornlerdpison
- Centre of Excellence in Agricultural Innovation for Graduate Entrepreneur and Faculty of Fisheries Technology and Aquatic Resources, Maejo University, Chiang Mai 50290, Thailand;
| | - Metee Jinakote
- School of Human Kinetics and Health, Faculty of Health Science Technology, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok 10210, Thailand;
| | - Chutima Srimaroeng
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (O.B.); (T.P.)
- Correspondence: ; Tel.: +66-53-935-362; Fax: +66-53-935-365
| |
Collapse
|
16
|
Is metformin a geroprotector? A peek into the current clinical and experimental data. Mech Ageing Dev 2020; 191:111350. [DOI: 10.1016/j.mad.2020.111350] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/25/2020] [Accepted: 09/01/2020] [Indexed: 02/08/2023]
|
17
|
Lee CH, Han JH, Kim S, Lee H, Kim S, Nam DH, Cho DH, Woo CH. Metformin ameliorates bile duct ligation-induced acute hepatic injury via regulation of ER stress. BMB Rep 2020. [PMID: 31791444 PMCID: PMC7330811 DOI: 10.5483/bmbrep.2020.53.6.169] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Cholestasis is a condition in which the bile duct becomes narrowed or clogged by a variety of factors and bile acid is not released smoothly. Bile acid-induced liver injury is facilitated by necrotic cell death, neutrophil infiltration, and inflammation. Metformin, the first-line treatment for type 2 diabetes, is known to reduce not only blood glucose but also inflammatory responses. In this study, we investigated the effects of metformin on liver injury caused by cholestasis with bile acid-induced hepatocyte injury. Static bile acid-induced liver injury is thought to be related to endoplasmic reticulum (ER) stress, inflammatory response, and chemokine expression. Metformin treatment reduced liver injury caused by bile acid, and it suppressed ER stress, inflammation, chemokine expression, and neutrophil infiltration. Similar results were obtained in mouse primary hepatocytes exposed to bile acid. Hepatocytes treated with tauroursodeoxycholic acid, an ER stress inhibitor, showed inhibition of ER stress, as well as reduced levels of inflammation and cell death. These results suggest that metformin may protect against liver injury by suppressing ER stress and inflammation and reducing chemokine expression.
Collapse
Affiliation(s)
- Chi-Ho Lee
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Jung-Hwa Han
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Sujin Kim
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Heejung Lee
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Suji Kim
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Dae-Hwan Nam
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Du-Hyong Cho
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| | - Chang-Hoon Woo
- Department of Pharmacology and Smart-Ageing Convergence Research Center, Yeungnam University College of Medicine, Daegu 42415, Korea
| |
Collapse
|
18
|
Jacob S, Knoll S, Huhn C, Köhler HR, Tisler S, Zwiener C, Triebskorn R. Effects of guanylurea, the transformation product of the antidiabetic drug metformin, on the health of brown trout ( Salmo trutta f. fario). PeerJ 2019; 7:e7289. [PMID: 31338260 PMCID: PMC6626654 DOI: 10.7717/peerj.7289] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/12/2019] [Indexed: 12/30/2022] Open
Abstract
Background Guanylurea is the main transformation product of the antidiabetic drug metformin, which is one of the most prescribed pharmaceuticals worldwide. Due to the high rate of microbial degradation of metformin in sewage treatment plants, guanylurea occurs in higher concentrations in surface waters than its parent compound and could therefore affect aquatic wildlife. In this context, data for fish are scarce up to now which made us investigate the health of brown trout (Salmo trutta f. fario) in response to guanylurea. Methods In two experiments, eggs plus developing larvae and juvenile brown trout were exposed to three different concentrations of guanylurea (10, 100 and 1,000 µg/L) and, as a negative control, filtered tap water without this compound. Low internal concentrations were determined. The investigated parameters were mortality, length, weight, condition factor, tissue integrity of the liver and kidney, levels of stress proteins and lipid peroxides, as well as behavioural and developmental endpoints. It was found that guanylurea did not significantly change any of these parameters in the tested concentration range. Results In conclusion, these results do not give rise to concern that guanylurea could negatively affect the health or the development of brown trout under field conditions. Nevertheless, more studies focusing on further parameters and other species are highly needed for a more profound environmental risk assessment of guanylurea.
Collapse
Affiliation(s)
- Stefanie Jacob
- University of Tübingen, Animal Physiological Ecology, Tübingen, Germany
| | - Sarah Knoll
- University of Tübingen, Effect-based Environmental Analysis, Tübingen, Germany
| | - Carolin Huhn
- University of Tübingen, Effect-based Environmental Analysis, Tübingen, Germany
| | - Heinz-R Köhler
- University of Tübingen, Animal Physiological Ecology, Tübingen, Germany
| | - Selina Tisler
- University of Tübingen, Environmental Analytical Chemistry, Tübingen, Germany
| | - Christian Zwiener
- University of Tübingen, Environmental Analytical Chemistry, Tübingen, Germany
| | - Rita Triebskorn
- University of Tübingen, Animal Physiological Ecology, Tübingen, Germany.,Steinbeis Transfer Center for Ecotoxicology and Ecophysiology, Rottenburg, Germany
| |
Collapse
|
19
|
Hepatoprotective activity of metformin: A new mission for an old drug? Eur J Pharmacol 2019; 850:1-7. [PMID: 30753869 DOI: 10.1016/j.ejphar.2019.02.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 02/02/2019] [Accepted: 02/08/2019] [Indexed: 02/06/2023]
Abstract
Metformin, as a dimethyl biguanide prescribed as the first-line medication for treatment of type 2 diabetes mellitus, is one of the most frequently used drugs, worldwide. However, the beneficial effects of metformin are not limited to insulin sensitizing and blood glucose lowering effects as recent clinical trials deciphered lower cancer risk in metformin users. In addition, metformin protected the liver against chemical or viral hepatotoxicants through various mechanisms including activation of AMPK via inhibition of mitochondrial complex I, inhibition of mitogen activated protein kinase (MAPK) and inhibition of Smads phosphorylation. Clinical trials are under way to assess possible additive effects of metformin when co-administered along with the standard regimen for hepatocellular carcinoma (HCC) treatment. This review outlines the molecular mechanisms behind protective activity of metformin against different liver diseases.
Collapse
|
20
|
Dogan Turacli I, Candar T, Yuksel EB, Kalay S, Oguz AK, Demirtas S. Potential effects of metformin in DNA BER system based on oxidative status in type 2 diabetes. Biochimie 2018; 154:62-68. [PMID: 30098371 DOI: 10.1016/j.biochi.2018.08.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/06/2018] [Indexed: 01/12/2023]
Abstract
Metformin is used to reduce hyperglycemia that induces energetic stress and leads to reduction in gluconeogenesis. Also, metformin inhibits complex I in oxidative phosphorylation, thereby decreasing cellular ATP levels. Activation of AMPK by the reduced ATP levels can induce inhibition of reactive oxygen species (ROS) production and activate p53-mediated DNA repair. DNA polymerase-β and XRCC1 function to repair DNA damages in the BER (base excision repair) system. In type 2 diabetes patients, metformin can enhance AMPK activation therefore suppress oxidative stress. The changes on oxidative stress may alter p53's function and effect many cellular pathways such as; DNA repair. In our project we aim to understand the effects of metformin on p53 and DNA-BER system based on the oxidative status in type 2 diabetes patients. Oxidative and antioxidative capacity, catalase, SOD, GPx activities and, DNA pol beta, XRCC1 and p53 levels were measured in metformin using or non-using type 2 diabetes patients and controls. Metformin enhanced SOD and GPx activities in type 2 diabetes patients but the reflection of this increase to the total antioxidant capacity was not significant. Although the increase in DNA pol beta was not significant, XRCC1 and p53 levels were significantly upregulated with metformin treatment in type 2 diabetes patients. Our study reinforces the potential benefit of metformin in antioxidative capacity to protect cells from diabetic oxidative stress and in regulation of DNA BER system.
Collapse
Affiliation(s)
| | - Tuba Candar
- Medical Biochemistry Department, Ufuk University, Ankara, 06520, Turkey
| | | | - Sebnem Kalay
- Internal Medicine Department, Ufuk University, Ankara, 06520, Turkey
| | - Ali Kemal Oguz
- Internal Medicine Department, Ufuk University, Ankara, 06520, Turkey
| | - Selda Demirtas
- Medical Biochemistry Department, Ufuk University, Ankara, 06520, Turkey
| |
Collapse
|
21
|
Lin Y, Wang K, Ma C, Wang X, Gong Z, Zhang R, Zang D, Cheng Y. Evaluation of Metformin on Cognitive Improvement in Patients With Non-dementia Vascular Cognitive Impairment and Abnormal Glucose Metabolism. Front Aging Neurosci 2018; 10:227. [PMID: 30100873 PMCID: PMC6074058 DOI: 10.3389/fnagi.2018.00227] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
Abstract
Objective: Recent studies have suggested that metformin can penetrate the blood–brain barrier, protecting neurons via anti-inflammatory action and improvement of brain energy metabolism. In this study, we aim to investigate the effect of metformin on cognitive function in patients with abnormal glucose metabolism and non-dementia vascular cognitive impairment (NDVCI). Methods: One hundred patients with NDVCI and abnormal glucose metabolism were randomly allocated into two groups: metformin and donepezil (n = 50) or acarbose and donepezil (n = 50). The neuropsychological status, glucose metabolism, and common carotid arteries intima–media thickness (CCA-IMT) before and after a year of treatment, were measured and compared between the groups. Results: Ninety four patients completed all the assessment and follow-up. After a year of treatment, there was a decrease in Alzheimer’s disease Assessment Scale-Cognitive Subscale scores and the duration of the Trail Making Test in the metformin-donepezil group. Furthermore, these patients showed a significant increase in World Health Organization–University of California–Los Angeles Auditory Verbal Learning Test scores after treatment (all P < 0.05). However, there was no obvious improvement in cognitive function in the acarbose-donepezil group. We also observed a significant decrease in the level of fasting insulin and insulin resistance (IR) index in the metformin-donepezil group, with a lower CCA-IMT value than that in the acarbose-donepezil group after a year of treatment (P < 0.05). Conclusion: We conclude that metformin can improve cognitive function in patients with NDVCI and abnormal glucose metabolism, especially in terms of performance function. Improved cognitive function may be related to improvement of IR and the attenuated progression of IMT. Trial Registration:ChiCTR-IPR-17011855.
Collapse
Affiliation(s)
- Yufeng Lin
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Kaiyuan Wang
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chunchao Ma
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Xuesong Wang
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Zhongying Gong
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Rui Zhang
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Dawei Zang
- Department of Neurology, Tianjin First Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Yan Cheng
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
22
|
Xing P, Feng Y, Niu Y, Li Q, Zhang Z, Dong L, Wang C. A Water-Soluble, Two-Photon Probe for Imaging Endogenous Hypochlorous Acid in Live Tissue. Chemistry 2018; 24:5748-5753. [DOI: 10.1002/chem.201800249] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Panfei Xing
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Yanxian Feng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Yiming Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Qiu Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Zhe Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology; Nanjing University Institution; Nanjing 210093 P. R. China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences; University of Macau; Avenida da Universidade Macau SAR P. R. China
| |
Collapse
|
23
|
Najafi M, Cheki M, Rezapoor S, Geraily G, Motevaseli E, Carnovale C, Clementi E, Shirazi A. Metformin: Prevention of genomic instability and cancer: A review. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 827:1-8. [PMID: 29502733 DOI: 10.1016/j.mrgentox.2018.01.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/28/2017] [Accepted: 01/15/2018] [Indexed: 12/21/2022]
Abstract
The diabetes drug metformin can mitigate the genotoxic effects of cytotoxic agents and has been proposed to prevent or even cure certain cancers. Metformin reduces DNA damage by mechanisms that are only incompletely understood. Metformin scavenges free radicals, including reactive oxygen species and nitric oxide, which are produced by genotoxicants such as ionizing or non-ionizing radiation, heavy metals, and chemotherapeutic agents. The drug may also increase the activities of antioxidant enzymes and inhibit NADPH oxidase, cyclooxygenase-2, and inducible nitric oxide synthase, thereby limiting macrophage recruitment and inflammatory responses. Metformin stimulates the DNA damage response (DDR) in the homologous end-joining, homologous recombination, and nucleotide excision repair pathways. This review focuses on the protective properties of metformin against genomic instability.
Collapse
Affiliation(s)
- Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Mohsen Cheki
- Department of Radiologic Technology, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Saeed Rezapoor
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazale Geraily
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Motevaseli
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Carla Carnovale
- Department of Biomedical and Clinical Sciences L. Sacco, Unit of Clinical Pharmacology, ASST Fatebenefratelli-Sacco University Hospital, Università di Milano, Milan, Italy
| | - Emilio Clementi
- Scientific Institute, IRCCS E. Medea, Bosisio Parini, Lecco, Italy; Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, Consiglio Nazionale delle Ricerche Institute of Neuroscience, L. Sacco University Hospital, Università di Milano, Milan, Italy
| | - Alireza Shirazi
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Trilla-Fuertes L, Gámez-Pozo A, Arevalillo JM, Díaz-Almirón M, Prado-Vázquez G, Zapater-Moros A, Navarro H, Aras-López R, Dapía I, López-Vacas R, Nanni P, Llorente-Armijo S, Arias P, Borobia AM, Maín P, Feliú J, Espinosa E, Fresno Vara JÁ. Molecular characterization of breast cancer cell response to metabolic drugs. Oncotarget 2018. [PMID: 29515760 PMCID: PMC5839391 DOI: 10.18632/oncotarget.24047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Metabolic reprogramming is a hallmark of cancer. It has been described that breast cancer subtypes present metabolism differences and this fact enables the possibility of using metabolic inhibitors as targeted drugs in specific scenarios. In this study, breast cancer cell lines were treated with metformin and rapamycin, showing a heterogeneous response to treatment and leading to cell cycle disruption. The genetic causes and molecular effects of this differential response were characterized by means of SNP genotyping and mass spectrometry-based proteomics. Protein expression was analyzed using probabilistic graphical models, showing that treatments elicit various responses in some biological processes such as transcription. Moreover, flux balance analysis using protein expression values showed that predicted growth rates were comparable with cell viability measurements and suggesting an increase in reactive oxygen species response enzymes due to metformin treatment. In addition, a method to assess flux differences in whole pathways was proposed. Our results show that these diverse approaches provide complementary information and allow us to suggest hypotheses about the response to drugs that target metabolism and their mechanisms of action.
Collapse
Affiliation(s)
- Lucía Trilla-Fuertes
- Molecular Oncology and Pathology Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain.,Biomedica Molecular Medicine SL, Madrid, Spain
| | - Angelo Gámez-Pozo
- Molecular Oncology and Pathology Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain.,Biomedica Molecular Medicine SL, Madrid, Spain
| | - Jorge M Arevalillo
- Operational Research and Numerical Analysis, National Distance Education University (UNED), Madrid, Spain
| | | | - Guillermo Prado-Vázquez
- Molecular Oncology and Pathology Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | - Andrea Zapater-Moros
- Molecular Oncology and Pathology Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | - Hilario Navarro
- Operational Research and Numerical Analysis, National Distance Education University (UNED), Madrid, Spain
| | - Rosa Aras-López
- Congenital Malformations Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Irene Dapía
- Pharmacogenetics Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Autonomous University of Madrid, Madrid, Spain.,Biomedical Research Networking Center on Rare Diseases-CIBERER, ISCIII, Madrid, Spain
| | - Rocío López-Vacas
- Molecular Oncology and Pathology Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | - Paolo Nanni
- Functional Genomics Center Zurich, University of Zurich/ETH Zurich, Zurich, Switzerland
| | - Sara Llorente-Armijo
- Molecular Oncology and Pathology Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain
| | - Pedro Arias
- Pharmacogenetics Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Autonomous University of Madrid, Madrid, Spain.,Biomedical Research Networking Center on Rare Diseases-CIBERER, ISCIII, Madrid, Spain
| | - Alberto M Borobia
- Clinical Pharmacology Department, La Paz University Hospital School of Medicine, IdiPAZ, Autonomous University of Madrid, Madrid, Spain
| | - Paloma Maín
- Department of Statistics and Operations Research, Faculty of Mathematics, Complutense University of Madrid, Madrid, Spain
| | - Jaime Feliú
- Medical Oncology Service, La Paz University Hospital-IdiPAZ, Madrid, Spain.,Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain.,Cátedra UAM-AMGEN, Universidad Autónoma de Madrid, Madrid, Spain
| | - Enrique Espinosa
- Medical Oncology Service, La Paz University Hospital-IdiPAZ, Madrid, Spain.,Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| | - Juan Ángel Fresno Vara
- Molecular Oncology and Pathology Lab, Institute of Medical and Molecular Genetics-INGEMM, La Paz University Hospital-IdiPAZ, Madrid, Spain.,Biomedica Molecular Medicine SL, Madrid, Spain.,Biomedical Research Networking Center on Oncology-CIBERONC, ISCIII, Madrid, Spain
| |
Collapse
|
25
|
Markowicz-Piasecka M, Sikora J, Szydłowska A, Skupień A, Mikiciuk-Olasik E, Huttunen KM. Metformin - a Future Therapy for Neurodegenerative Diseases : Theme: Drug Discovery, Development and Delivery in Alzheimer's Disease Guest Editor: Davide Brambilla. Pharm Res 2017; 34:2614-2627. [PMID: 28589443 PMCID: PMC5736777 DOI: 10.1007/s11095-017-2199-y] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 05/30/2017] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a complex, chronic and progressive metabolic disease, which is characterized by relative insulin deficiency, insulin resistance, and high glucose levels in blood. Esteemed published articles and epidemiological data exhibit an increased risk of developing Alzheimer's disease (AD) in diabetic pateints. Metformin is the most frequently used oral anti-diabetic drug, which apart from hypoglycaemic activity, improves serum lipid profiles, positively influences the process of haemostasis, and possesses anti-inflammatory properties. Recently, scientists have put their efforts in establishing metformin's role in the treatment of neurodegenerative diseases, such as AD, amnestic mild cognitive impairment and Parkinson's disease. Results of several clinical studies confirm that long term use of metformin in diabetic patients contributes to better cognitive function, compared to participants using other anti-diabetic drugs. The exact mechanism of metformin's advantageous activity in AD is not fully understood, but scientists claim that activation of AMPK-dependent pathways in human neural stem cells might be responsible for the neuroprotective activity of metformin. Metformin was also found to markedly decease Beta-secretase 1 (BACE1) protein expression and activity in cell culture models and in vivo, thereby reducing BACE1 cleavage products and the production of Aβ (β-amyloid). Furthermore, there is also some evidence that metformin decreases the activity of acetylcholinesterase (AChE), which is responsible for the degradation of acetylcholine (Ach), a neurotransmitter involved in the process of learning and memory. In regard to the beneficial effects of metformin, its anti-inflammatory and anti-oxidative properties cannot be omitted. Numerous in vitro and in vivo studies have confirmed that metformin ameliorates oxidative damage.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry Drug Analysis and Radiopharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry Drug Analysis and Radiopharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Aleksandra Szydłowska
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry Drug Analysis and Radiopharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Agata Skupień
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry Drug Analysis and Radiopharmacy, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland
| | - Elżbieta Mikiciuk-Olasik
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul., Muszyńskiego 1, 90-151, Lodz, Poland
| | - Kristiina M. Huttunen
- School Of Pharmacy, Faculty of Health Sciences, University of Eastern Finland,, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland
| |
Collapse
|
26
|
Dayangan Sayan C, Karaca G, Sema Ozkan Z, Tulmac OB, Ceylan Isik A, Devrim T, Aydin G, Yeral I. What is the protective effect of metformin on rat ovary against ischemia-reperfusion injury? J Obstet Gynaecol Res 2017; 44:278-285. [DOI: 10.1111/jog.13524] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/17/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Cemile Dayangan Sayan
- Department of Obstetrics and Gynecology; Kırıkkale University Faculty of Medicine; Kırıkkale Turkey
| | - Gokhan Karaca
- Department of General Surgery; Kırıkkale University Faculty of Medicine; Kırıkkale Turkey
| | - Zehra Sema Ozkan
- Department of Obstetrics and Gynecology; Kırıkkale University Faculty of Medicine; Kırıkkale Turkey
| | - Ozlem B. Tulmac
- Department of Obstetrics and Gynecology; Kırıkkale University Faculty of Medicine; Kırıkkale Turkey
| | - Aslı Ceylan Isik
- Department of Pharmacology; Kırıkkale University Faculty of Medicine; Kırıkkale Turkey
| | - Tuba Devrim
- Department of Pathology; Kırıkkale University Faculty of Medicine; Kırıkkale Turkey
| | - Gülcin Aydin
- Department of Anesthesiology and Reanimation; Kırıkkale University Faculty of Medicine; Kırıkkale Turkey
| | - Ilkin Yeral
- Department of Obstetrics and Gynecology; Kırıkkale University Faculty of Medicine; Kırıkkale Turkey
| |
Collapse
|
27
|
Fan K, Wu K, Lin L, Ge P, Dai J, He X, Hu K, Zhang L. Metformin mitigates carbon tetrachloride-induced TGF-β1/Smad3 signaling and liver fibrosis in mice. Biomed Pharmacother 2017; 90:421-426. [DOI: 10.1016/j.biopha.2017.03.079] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/24/2017] [Accepted: 03/26/2017] [Indexed: 02/06/2023] Open
|
28
|
Karise I, Ornellas F, Barbosa-da-Silva S, Matsuura C, Del Sol M, Aguila MB, Mandarim-de-Lacerda CA. Liver and Metformin: Lessons of a fructose diet in mice. BIOCHIMIE OPEN 2017; 4:19-30. [PMID: 29450137 PMCID: PMC5801827 DOI: 10.1016/j.biopen.2017.01.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/27/2017] [Indexed: 12/11/2022]
Abstract
Studies show that the continuous consumption of fructose can lead to nonalcoholic fatty liver disease (NAFLD) and steatohepatitis. We aimed to investigate the role of Metformin in an animal model of liver injury caused by fructose intake, focusing on the molecular markers of lipogenesis, beta-oxidation, and antioxidant defenses. Male three months old C57BL/6 mice were divided into control group (C) and fructose group (F, 47% fructose), maintained for ten weeks. After, the groups received Metformin or vehicle for a further eight weeks: control (C), control + Metformin (CM), fructose (F), and fructose + Metformin (FM). Fructose resulted in hepatic steatosis, insulin resistance and lower insulin sensitivity in association with higher mRNA levels of proteins linked with de novo lipogenesis and increased lipid peroxidation. Fructose diminished mRNA expression of antioxidant enzymes, and of proteins responsible for mitochondrial biogenesis. Metformin reduced de novo lipogenesis and increased the expression of proteins related to mitochondrial biogenesis, thereby increasing beta-oxidation and decreasing lipid peroxidation. Also, Metformin upregulated the expression and activity of antioxidant enzymes, providing a defense against increased reactive oxygen species generation. Therefore, a significant reduction in triglyceride accumulation in the liver, steatosis and lipid peroxidation was observed in the FM group. In conclusion, fructose increases de novo lipogenesis, reduces the antioxidant defenses, and diminishes mitochondrial biogenesis. After an extended period of fructose intake, Metformin treatment, even in continuing the fructose intake, can reverse, at least partially, the liver injury and prevents NAFLD progression to more severe states. Fructose increases lipogenesis and lipid peroxidation, reduces the antioxidant defenses, and mitochondrial biogenesis. Metformin mechanism of action remains partially understood and controversial. Metformin can reverse the liver injury preventing the progression to more severe states.
Collapse
Affiliation(s)
- Iara Karise
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Fernanda Ornellas
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Sandra Barbosa-da-Silva
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Cristiane Matsuura
- Laboratory of Membrane Transport, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Mariano Del Sol
- Doctoral Programing on Morphological Sciences, Universidad de La Frontera, Temuco, Chile
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil.,Doctoral Programing on Morphological Sciences, Universidad de La Frontera, Temuco, Chile
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil.,Doctoral Programing on Morphological Sciences, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
29
|
Saeedi Saravi SS, Hasanvand A, Shahkarami K, Dehpour AR. The protective potential of metformin against acetaminophen-induced hepatotoxicity in BALB/C mice. PHARMACEUTICAL BIOLOGY 2016; 54:2830-2837. [PMID: 27252117 DOI: 10.1080/13880209.2016.1185633] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 04/30/2016] [Indexed: 06/05/2023]
Abstract
CONTEXT Acetaminophen overdose is regarded to a common cause of acute liver failure. The hepatotoxicity leads to mitochondrial oxidative stress and subsequent necrotic hepatocellular death. OBJECTIVE This study examines the protective effect of metformin on acetaminophen-induced oxidative stress, inflammation and subsequent hepatotoxicity in mice. MATERIALS AND METHODS Male BALB/c mice were orally administered to acetaminophen (250 mg/kg/d) for a 7-day period. The mice received metformin (100 and 200 mg/kg/d, p.o.) for 21 days. To evaluate acetaminophen-induced oxidative stress, liver tissue level of malodialdehyde (MDA), end product of membrane lipid peroxidation, and activities of superoxide dismutase (SOD) and glutathione (GSH) were measured. Histological analysis and measurement of serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP) were performed. Moreover, tissue concentrations of proinflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), along with, C-reactive protein (CRP) were assessed. RESULTS Acetaminophen caused focal hepatocyte necrosis, inflammation and fatty degeneration, as well as increased tissue levels of AST, ALT, ALP and MDA, and also decreased GSH and SOD activities. Moreover, IL-6, TNF-α and CRP levels were increased following acetaminophen hepatotoxicity. Metformin (200 mg/kg/d) significantly normalized MDA, SOD and GSH levels (p < 0.001), and exerted a hepatoprotective effect by significant decreasing ALT, AST and ALP concentrations (p < 0.001). The tissue levels of IL-6, TNF-α and CRP were markedly decreased by 21-day treatment with metformin (200 mg/kg/d) (p < 0.001). DISCUSSION The results suggest metformin protects hepatocytes against acute acetaminophen toxicity. Metformin is indicated to diminish oxidative stress, proinflammatory cytokines, and hepatocyte necrosis.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- a Department of Pharmacology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran
- b Experimental Medicine Research Center, Tehran University of Medical Sciences , Tehran , Iran
- c Department of Toxicology-Pharmacology, Faculty of Pharmacy , Gilan University of Medical Sciences , Rasht , Iran
| | - Amin Hasanvand
- a Department of Pharmacology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran
- d Department of Pharmacology and Toxicology, Faculty of Pharmacy , Lorestan University of Medical Sciences , Khorramabad , Iran
| | - Kourosh Shahkarami
- e Department of Neuroscience and Addiction, School of Advanced Technologies in Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Ahmad Reza Dehpour
- a Department of Pharmacology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran
- b Experimental Medicine Research Center, Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
30
|
Cheki M, Shirazi A, Mahmoudzadeh A, Bazzaz JT, Hosseinimehr SJ. The radioprotective effect of metformin against cytotoxicity and genotoxicity induced by ionizing radiation in cultured human blood lymphocytes. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2016; 809:24-32. [PMID: 27692296 DOI: 10.1016/j.mrgentox.2016.09.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 08/23/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022]
Abstract
Metformin is a widely prescribed drug used in the treatment of patients with type 2 diabetes. In this study, the radioprotective effect of metformin was investigated against cytotoxicity and genotoxicity induced by ionizing radiation (IR) in human peripheral blood lymphocytes. Human lymphocytes were treated with metformin at concentrations 10 and 50μM for 2h and irradiated with 6MV X-rays. The radiation antagonistic potential of metformin was assessed by MTT [3-(4,5-dimethyl-2-thiaozolyl)-2,5-diphenyl-2H tetrazolium bromide] assay, chromosomal aberration (CA) analysis, cytokinesis blocked micronucleus (CBMN) assay, and flow cytometry. Observations demonstrated a radiation-dose-dependent decrease in the percentage of cell viability after 24h. It was found that pretreatment with metformin (10 and 50μM) increased the percentage of cell viability. A highly significant dose modifying factor (DMF) 1.35 and 1.42 was observed for 10 and 50μM metformin, respectively. Metformin (10 and 50μM) pretreatment significantly decreased the frequency of dicentrics (DCs), acentric fragments (AFs), rings (RIs), micronuclei (MN), and nucleoplasmic bridges (NPBs) in irradiated human peripheral blood lymphocytes. Also, treatment with metformin (10 and 50μM) without irradiation did not increase the number of MN, NPBs, DCs, AFs, RIs, and did not show a cytostatic effect in the human peripheral blood lymphocytes. On the other hand, metformin treatment (10 and 50μM) 2h prior to irradiation significantly reduced X-radiation-induced apoptotic incidence in human lymphocytes. The present study demonstrates metformin to be an effective radioprotector against DNA damage and apoptosis induced by IR in human lymphocytes. These data have an important application for the protection of lymphocytes from the genetic damage and side-effects induced by radiotherapy in cancer patients.
Collapse
Affiliation(s)
- Mohsen Cheki
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences and Health Services, Tehran, Iran
| | - Alireza Shirazi
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences and Health Services, Tehran, Iran.
| | - Aziz Mahmoudzadeh
- Department of Biosciences and Biotechnology, Malek-Ashtar University of Technology, Tehran, Iran
| | - Javad Tavakkoly Bazzaz
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences and Health Services, Tehran, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
31
|
Mummidi S, Das NA, Carpenter AJ, Kandikattu H, Krenz M, Siebenlist U, Valente AJ, Chandrasekar B. Metformin inhibits aldosterone-induced cardiac fibroblast activation, migration and proliferation in vitro, and reverses aldosterone+salt-induced cardiac fibrosis in vivo. J Mol Cell Cardiol 2016; 98:95-102. [PMID: 27423273 DOI: 10.1016/j.yjmcc.2016.07.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/08/2016] [Accepted: 07/13/2016] [Indexed: 11/29/2022]
Abstract
The overall goals of this study were to investigate whether metformin exerts anti-fibrotic effects in aldosterone (Aldo)+salt-treated wild type mouse hearts, and determine the underlying molecular mechanisms in isolated adult cardiac fibroblasts (CF). In vitro, Aldo induced CF activation, migration, and proliferation, and these effects were inhibited by metformin. Further, Aldo induced PPM1A (Protein Phosphatase Magnesium Dependent 1A) activation and inhibited AMPK phosphorylation. At a pharmacologically relevant concentration, metformin restored AMPK activation, and inhibited Aldo-induced Nox4/H2O2-dependent TRAF3IP2 induction, pro-inflammatory cytokine expression, and CF migration and proliferation. Further, metformin potentiated the inhibitory effects of spironolactone, a mineralocorticoid receptor antagonist, on Aldo-induced collagen expression, and CF migration and proliferation. These results were recapitulated in vivo, where metformin reversed Aldo+salt-induced oxidative stress, suppression of AMPK activation, TRAF3IP2 induction, pro-inflammatory cytokine expression, and cardiac fibrosis, without significantly modulating systolic blood pressure. These in vitro and in vivo data indicate that metformin has the potential to reduce adverse cardiac remodeling in hypertensive heart disease.
Collapse
Affiliation(s)
- Srinivas Mummidi
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Nitin A Das
- Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Andrea J Carpenter
- Department of Cardiothoracic Surgery, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | | | - Maike Krenz
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, MO 65211, USA
| | | | - Anthony J Valente
- Department of Medicine, University of Texas Health Science Center, San Antonio, TX 78229, USA
| | - Bysani Chandrasekar
- Medicine/Cardiology, University of Missouri School of Medicine, Columbia, MO 65211, USA; Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, MO 65201, USA.
| |
Collapse
|
32
|
García-Heredia A, Riera-Borrull M, Fort-Gallifa I, Luciano-Mateo F, Cabré N, Hernández-Aguilera A, Joven J, Camps J. Metformin administration induces hepatotoxic effects in paraoxonase-1-deficient mice. Chem Biol Interact 2016; 249:56-63. [PMID: 26945512 DOI: 10.1016/j.cbi.2016.03.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/23/2016] [Accepted: 03/01/2016] [Indexed: 12/11/2022]
Abstract
Metformin is the first-line pharmacological treatment of diabetes. In these patients, metformin reduces body weight and decreases the risk of diabetes-related complications such as cardiovascular disease. However, whether metformin elicits beneficial effects on liver histology is a controversial issue and, as yet, there is no consensus. Paraoxonase-1 (PON1), an enzyme synthesized mainly by the liver, degrades lipid peroxides and reduces oxidative stress. PON1 activities are decreased in chronic liver diseases. We evaluated the effects of metformin in the liver of PON1-deficient mice which, untreated, present a mild degree of liver steatosis. Metformin administration aggravated inflammation in animals given a standard mouse chow and in those fed a high-fat diet. Also, it was associated with a higher degree of steatosis in animals fed a standard chow diet. This report is a cautionary note regarding the prescription of metformin for the treatment of diabetes in patients with concomitant liver impairment.
Collapse
Affiliation(s)
- Anabel García-Heredia
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Marta Riera-Borrull
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Isabel Fort-Gallifa
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain; Laboratori de Referència Sud, Hospital Universitari de Sant Joan, Reus, Spain
| | - Fedra Luciano-Mateo
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Noemí Cabré
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Anna Hernández-Aguilera
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Jorge Joven
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Jordi Camps
- Unitat de Recerca Biomèdica (CRB-URB), Hospital Universitari de Sant Joan, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, Reus, Spain.
| |
Collapse
|
33
|
Khallaghi B, Safarian F, Nasoohi S, Ahmadiani A, Dargahi L. Metformin-induced protection against oxidative stress is associated with AKT/mTOR restoration in PC12 cells. Life Sci 2016; 148:286-92. [DOI: 10.1016/j.lfs.2016.02.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 01/26/2016] [Accepted: 02/07/2016] [Indexed: 12/21/2022]
|
34
|
Is Liver Enzyme Release Really Associated with Cell Necrosis Induced by Oxidant Stress? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3529149. [PMID: 26798419 PMCID: PMC4699024 DOI: 10.1155/2016/3529149] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 10/11/2015] [Indexed: 12/16/2022]
Abstract
Hepatic diseases are a major concern worldwide. Increased specific plasma enzyme activities are considered diagnostic features for liver diseases, since enzymes are released into the blood compartment following the deterioration of the organ. Release of liver mitochondrial enzymes is considered strong evidence for hepatic necrosis, which is associated with an increased production of ROS, often leading to greater hepatic lipid peroxidation. Lipotoxic mediators and intracellular signals activated Kupffer cells, which provides evidence strongly suggesting the participation of oxidant stress in acute liver damage, inducing the progression of liver injury to chronic liver damage. Elevated transaminase activities are considered as an index marker of hepatotoxicity, linked to oxidant stress. However, a drastic increase of serum activities of liver enzyme markers ought not necessarily to reflect liver cell death. In fact, increased serum levels of cytoplasmic enzymes have readily been observed after partial hepatectomy (PH) in the regenerating liver of rats. In this regard, we are now showing that in vitro modifications of the oxidant status affect differentially the release of liver enzymes, indicating that this release is a strictly controlled event and not directly related to the onset of oxidant stress of the liver.
Collapse
|
35
|
Cai L, Hu K, Lin L, Ai Q, Ge P, Liu Y, Dai J, Ye B, Zhang L. AMPK dependent protective effects of metformin on tumor necrosis factor-induced apoptotic liver injury. Biochem Biophys Res Commun 2015; 465:381-6. [DOI: 10.1016/j.bbrc.2015.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 08/03/2015] [Indexed: 12/14/2022]
|
36
|
Nguyen TMD, Seigneurin F, Froment P, Combarnous Y, Blesbois E. The 5'-AMP-Activated Protein Kinase (AMPK) Is Involved in the Augmentation of Antioxidant Defenses in Cryopreserved Chicken Sperm. PLoS One 2015. [PMID: 26222070 PMCID: PMC4519194 DOI: 10.1371/journal.pone.0134420] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Semen cryopreservation is a unique tool for the management of animal genetic diversity. However, the freeze-thaw process causes biochemical and physical alterations which make difficult the restoration of sperm energy-dependent functions needed for fertilization. 5'-AMP activated protein kinase (AMPK) is a key sensor and regulator of intracellular energy metabolism. Mitochondria functions are known to be severely affected during sperm cryopreservation with deleterious oxidative and peroxidative effects leading to cell integrity and functions damages. The aim of this study was thus to examine the role of AMPK on the peroxidation/antioxidant enzymes defense system in frozen-thawed sperm and its consequences on sperm functions. Chicken semen was diluted in media supplemented with or without AMPK activators (AICAR or Metformin [MET]) or inhibitor (Compound C [CC]) and then cryopreserved. AMPKα phosphorylation, antioxidant enzymes activities, mitochondrial potential, ATP, citrate, viability, acrosome reaction ability (AR) and various motility parameters were negatively affected by the freeze-thaw process while reactive oxygen species (ROS) production, lipid peroxidation (LPO) and lactate concentration were dramatically increased. AICAR partially restored superoxide dismutase (SOD), Glutathione Peroxidase (GPx) and Glutathione Reductase (GR), increased ATP, citrate, and lactate concentration and subsequently decreased the ROS and LPO (malondialdehyde) in frozen-thawed semen. Motility parameters were increased (i.e., + 23% for motility, + 34% for rapid sperm) as well as AR (+ 100%). MET had similar effects as AICAR except that catalase activity was restored and that ATP and mitochondrial potential were further decreased. CC showed effects opposite to AICAR on SOD, ROS, LPO and AR and motility parameters. Taken together, our results strongly suggest that, upon freeze-thaw process, AMPK stimulated intracellular anti-oxidative defense enzymes through ATP regulation, thus reducing ROS and lipid peroxidation, and consequently partially restoring several essential sperm functions and leading to a better quality of cryopreserved sperm.
Collapse
Affiliation(s)
- Thi Mong Diep Nguyen
- INRA-CNRS, Unité Mixte de Recherche de Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
- Université François Rabelais, Tours, France
| | | | - Pascal Froment
- INRA-CNRS, Unité Mixte de Recherche de Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
| | - Yves Combarnous
- INRA-CNRS, Unité Mixte de Recherche de Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
| | - Elisabeth Blesbois
- INRA-CNRS, Unité Mixte de Recherche de Physiologie de la Reproduction et des Comportements, F-37380 Nouzilly, France
- * E-mail:
| |
Collapse
|
37
|
Wang J, Miao M, Zhang Y, Liu R, Li X, Cui Y, Qu L. Quercetin ameliorates liver injury induced with Tripterygium glycosides by reducing oxidative stress and inflammation. Can J Physiol Pharmacol 2015; 93:427-33. [PMID: 25894526 DOI: 10.1139/cjpp-2015-0038] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Quercetin (Que) is one of main compounds in Lysimachia christinae Hance (Christina loosestrife), and has both medicinal and nutritional value. Glycosides from Tripterygium wilfordii Hook.f. (léi gōng téng [the thunder duke vine]; TG) have diverse and broad bioactivities but with a high incidence of liver injury. Our previous study reported on the hepatoprotective properties of an ethanol extract from L. christinae against TG-induced liver injury in mice. This research is designed to observe, for the first time, the possible protective properties of the compound Que against TG-induced liver injury, and the underlying mechanisms that are involved in oxidative stress and anti-inflammation. The results indicated that TG caused excessive elevation in serum levels of alanine/aspartate transaminase (ALT/AST), alkaline phosphatase (ALP), gamma glutamyl transferase (γ-GT), and pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-α), as well as hepatic lipid peroxidation (all P < 0.01). On the other hand, following TG exposure, we observed significantly reduced levels of biomarkers, including hepatic glutathione (GSH), glutathione-S-transferase (GST), glutathione peroxidase (GPx), and the anti-inflammatory cytokine interleukin (IL)-10, as well as the enzyme activity and mRNA expression of copper- and zinc-containing superoxide dismutase (CuZn-SOD) and catalase (CAT) (all P < 0.01). Nevertheless, all of these alterations were reversed by the pre-administration of Que or the drug bifendate (positive control) for 7 consecutive days. Therefore, this study suggests that Que ameliorates TG-induced acute liver injury, probably through its ability to reduce oxidative stress and its anti-inflammatory properties.
Collapse
Affiliation(s)
- Junming Wang
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment & Chinese Medicine Development of Henan Province, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China., College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou 450001, China
| | | | | | | | | | | | | |
Collapse
|
38
|
Chen T, Gao J, Xiang P, Chen Y, Ji J, Xie P, Wu H, Xiao W, Wei Y, Wang S, Lan L, Ji H, Yan T. Protective effect of platycodin D on liver injury in alloxan-induced diabetic mice via regulation of Treg/Th17 balance. Int Immunopharmacol 2015; 26:338-48. [PMID: 25887267 DOI: 10.1016/j.intimp.2015.04.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 03/29/2015] [Accepted: 04/02/2015] [Indexed: 02/06/2023]
Abstract
Platycodin D is a major pharmacological constituent of Platycodi Radix with immunomodulatory activity. The present study was designed to investigate how platycodin D (PLD) reveals liver injury in diabetic mice and its mechanism. Fifty mice were divided into five groups randomly: control group, model group, rosiglitazone (ROG, 10 mg/kg) group, PLD (50 mg/kg) group, and PLD (100 mg/kg) group. Diabetes was induced with the injection of alloxan monohydrate (150 mg/kg) subcutaneously, and animals with blood glucose level of ≥250 mg/dl were considered as diabetic mice. After the first day of diabetes induction, the treatments were performed for 8 weeks. Then the animals were anaesthetized, and blood and liver samples were also collected for further assay. PLD significantly decreased the serum levels of glucose, insulin, interleukin-6 (IL-6), interleukin-1β, tumor necrosis factor-α (TNF-α), and interleukin (IL)-17A and increased IL-10 level in serum. PLD effectively downregulated aspartate transaminase (AST), alanine aminotransferase (ALT), total cholesterol (TC), and triglycerides (TG) in liver. PLD also attenuated liver histological change. In addition, PLD significantly attenuated IL-17A and IL-10 levels in vitro, flow cytometry (FCM) studies also showed that PLD remarkably inhibited Th17 cells and significantly increased Treg cells in liver tissues and spleen cells. Western blot demonstrated PLD inhibited the phosphorylation of JAK and STAT-3 and the expression of RORγt and increased the expression of Foxp3. The findings showed that PLD exerts beneficial effects on alloxan-induced liver injury in mice.
Collapse
Affiliation(s)
- Tong Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Jin Gao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Pengjun Xiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yongde Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Jing Ji
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Xie
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Wei Xiao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Yidan Wei
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Shumin Wang
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Li Lan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Hui Ji
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| | - Tianhua Yan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
39
|
Yang C, Gong X, Ai Q, Ge P, Lin L, Zhang L. 5-Aminoimidazole-4-carboxamide-1-β-d-ribofuranoside alleviated carbon tetrachloride-induced acute hepatitis in mice. Int Immunopharmacol 2015; 25:393-9. [PMID: 25711693 DOI: 10.1016/j.intimp.2015.02.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/28/2015] [Accepted: 02/11/2015] [Indexed: 01/02/2023]
Abstract
AMP-activated protein kinase (AMPK) is one of the principal cellular energy sensors participating in maintenance of energy balance but recent evidences also suggested that AMPK might be involved in the regulation of inflammation. In the present study, the AMPK activator 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) was used to investigate the potential roles of AMPK in carbon tetrachloride (CCl4)-induced acute hepatitis. The experimental data indicated that treatment with AICAR significantly decreased the elevation of plasma aminotransferases and alleviated hepatic histological abnormalities in CCl4-exposed mice. Treatment with AICAR also inhibited the increase of myeloperoxidase (MPO), the induction of TNF-α, IL-6, inducible nitric oxide synthase (iNOS), nitric oxide and the upregulation of matrix metalloproteinase 2 (MMP-2), MMP-3 and MMP-9 in mice exposed to CCl4. These effects were associated with suppressed nuclear accumulation of NF-κB p65. These results indicated that the AMPK activator AICAR effectively suppressed the inflammatory responses and alleviated liver damage induced by CCl4, implying that AMPK activation might be beneficial for ameliorating inflammation-based liver damage.
Collapse
Affiliation(s)
- Changming Yang
- Department of Anesthesiology, The First People's Hospital of Jingmen, Jingmen, Hubei Province, China
| | - Xianqiong Gong
- Hepatology Center, Xiamen Hospital of Traditional Chinese Medicine, Xiamen, Fujian Province, China
| | - Qing Ai
- Department of Physiology, Chongqing Medical University, Chongqing, China
| | - Pu Ge
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Ling Lin
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China.
| |
Collapse
|