51
|
Ahmed LA, Abdou FY, El Fiky AA, Shaaban EA, Ain-Shoka AA. Bradykinin-Potentiating Activity of a Gamma-Irradiated Bioactive Fraction Isolated from Scorpion (Leiurus quinquestriatus) Venom in Rats with Doxorubicin-Induced Acute Cardiotoxicity: Favorable Modulation of Oxidative Stress and Inflammatory, Fibrogenic and Apoptotic Pathways. Cardiovasc Toxicol 2021; 21:127-141. [PMID: 32860604 DOI: 10.1007/s12012-020-09602-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 08/18/2020] [Indexed: 10/23/2022]
Abstract
Although doxorubicin (Dox) is a backbone of chemotherapy, the search for an effective and safe therapy to revoke Dox-induced acute cardiotoxicity remains a critical matter in cardiology and oncology. The current study was the first to explore the probable protective effects of native and gamma-irradiated fractions with bradykinin-potentiating activity (BPA) isolated from scorpion (Leiurus quinquestriatus) venom against Dox-induced acute cardiotoxicity in rats. Native or irradiated fractions (1 μg/g) were administered intraperitoneally (i.p.) twice per week for 3 weeks, and Dox (15 mg/kg, i.p.) was administered on day 21 at 1 h after the last native or irradiated fraction treatment. Electrocardiographic (ECG) aberrations were ameliorated in the Dox-treated rats pretreated with the native fraction, and the irradiated fraction provided greater amelioration of ECG changes than that of the native fraction. The group pretreated with native protein with BPA also exhibited significant improvements in the levels of oxidative stress-related, inflammatory, angiogenic, fibrogenic, and apoptotic markers compared with those of the Dox group. Notably, the irradiated fraction restored these biomarkers to their normal levels. Additionally, the irradiated fraction ameliorated Dox-induced histological changes and alleviated the severity of cardiac injury to a greater extent than that of the native fraction. In conclusion, the gamma-irradiated detoxified fraction of scorpion venom elicited a better cardioprotective effect than that of the native fraction against Dox-induced acute cardiotoxicity in rats.
Collapse
Affiliation(s)
- Lamiaa A Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt.
| | - Fatma Y Abdou
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Atomic Energy Authority, Nasr, Cairo, Egypt
| | - Abir A El Fiky
- ANDI Center of Excellence in Antivenom Research, Vacsera, Egypt
| | - Esmat A Shaaban
- Department of Drug Radiation Research, National Center for Radiation Research and Technology, Atomic Energy Authority, Nasr, Cairo, Egypt
| | - Afaf A Ain-Shoka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt
| |
Collapse
|
52
|
Monahan DS, Almas T, Wyile R, Cheema FH, Duffy GP, Hameed A. Towards the use of localised delivery strategies to counteract cancer therapy-induced cardiotoxicities. Drug Deliv Transl Res 2021; 11:1924-1942. [PMID: 33449342 DOI: 10.1007/s13346-020-00885-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Cancer therapies have significantly improved cancer survival; however, these therapies can often result in undesired side effects to off target organs. Cardiac disease ranging from mild hypertension to heart failure can occur as a result of cancer therapies. This can warrant the discontinuation of cancer treatment in patients which can be detrimental, especially when the treatment is effective. There is an urgent need to mitigate cardiac disease that occurs as a result of cancer therapy. Delivery strategies such as the use of nanoparticles, hydrogels, and medical devices can be used to localise the treatment to the tumour and prevent off target side effects. This review summarises the advancements in localised delivery of anti-cancer therapies to tumours. It also examines the localised delivery of cardioprotectants to the heart for patients with systemic disease such as leukaemia where localised tumour delivery might not be an option.
Collapse
Affiliation(s)
- David S Monahan
- Anatomy & Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine Nursing and Health Science, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.,Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Talal Almas
- School of Medicine, RCSI University of Medicine and Health Sciences, 123, St. Stephens Green, Dublin 2, Dublin, D02 YN77, Ireland
| | - Robert Wyile
- Anatomy & Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine Nursing and Health Science, National University of Ireland Galway, Galway, Ireland
| | - Faisal H Cheema
- HCA Healthcare, Gulf Coast Division, Houston, TX, USA.,College of Medicine, University of Houston, Houston, TX, USA
| | - Garry P Duffy
- Anatomy & Regenerative Medicine Institute (REMEDI), School of Medicine, College of Medicine Nursing and Health Science, National University of Ireland Galway, Galway, Ireland.,Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland.,Tissue Engineering Research Group (TERG), Department of Anatomy, RCSI University of Medicine and Health Sciences, 123, St. Stephens Green, Dublin 2, Dublin, D02 YN77, Ireland.,Advanced Materials for Biomedical Engineering and Regenerative Medicine (AMBER), National University of Ireland, Trinity College Dublin &, Galway, Ireland.,Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
| | - Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy, RCSI University of Medicine and Health Sciences, 123, St. Stephens Green, Dublin 2, Dublin, D02 YN77, Ireland. .,Trinity Centre for Biomedical Engineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland.
| |
Collapse
|
53
|
Wu C, Hua K, Yang S, Tsai Y, Yang S, Hsieh C, Wu C, Chang J, Arbiser JL, Chang C, Chen A, Ka S. Tris DBA ameliorates IgA nephropathy by blunting the activating signal of NLRP3 inflammasome through SIRT1- and SIRT3-mediated autophagy induction. J Cell Mol Med 2020; 24:13609-13622. [PMID: 33135320 PMCID: PMC7753881 DOI: 10.1111/jcmm.15663] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 06/12/2020] [Accepted: 06/16/2020] [Indexed: 11/28/2022] Open
Abstract
Tris (dibenzylideneacetone) dipalladium (Tris DBA), a small-molecule palladium complex, can inhibit cell growth and proliferation in pancreatic cancer, lymphocytic leukaemia and multiple myeloma. Given that this compound is particularly active against B-cell malignancies, we have been suggested that it can alleviate immune complexes (ICs)-mediated conditions, especially IgA nephropathy (IgAN). The therapeutic effects of Tris DBA on glomerular cell proliferation and renal inflammation and mechanism of action were examined in a mouse model of IgAN. Treatment of IgAN mice with Tris DBA resulted in markedly improved renal function, albuminuria and renal pathology, including glomerular cell proliferation, neutrophil infiltration, sclerosis and periglomerular inflammation in the renal interstitium, together with (Clin J Am Soc Nephrol. 2011, 6, 1301-1307) reduced mitochondrial ROS generation; (Am J Physiol-Renal Physiol. 2011. 301, F1218-F1230) differentially regulated autophagy and NLRP3 inflammasome; (Clin J Am Soc Nephrol. 2012, 7, 427-436) inhibited phosphorylation of JNK, ERK and p38 MAPK signalling pathways, and priming signal of the NLRP3 inflammasome; and (Free Radic Biol Med. 2013, 61, 285-297) blunted NLRP3 inflammasome activation through SIRT1- and SIRT3-mediated autophagy induction, in renal tissues or cultured macrophages. In conclusion, Tris DBA effectively ameliorated the mouse IgAN model and targeted signalling pathways downstream of ICs-mediated interaction, which is a novel immunomodulatory strategy. Further development of Tris DBA as a therapeutic candidate for IgAN is warranted.
Collapse
Affiliation(s)
- Chung‐Yao Wu
- Graduate Institute of Life SciencesNational Defense Medical CenterTaipeiTaiwan
| | - Kuo‐Feng Hua
- Department of Biotechnology and Animal ScienceNational Ilan UniversityIlanTaiwan
| | - Shin‐Ruen Yang
- Graduate Institute of Life SciencesNational Defense Medical CenterTaipeiTaiwan
| | - Yi‐Shan Tsai
- Graduate Institute of Life SciencesNational Defense Medical CenterTaipeiTaiwan
| | - Shun‐Min Yang
- Department of PathologyTri‐Service General HospitalNational Defense Medical CenterTaipeiTaiwan
| | - Chih‐Yu Hsieh
- Department of Internal MedicineEn Chu Kong HospitalNew Taipei CityTaiwan
- Renal Care Joint FoundationNew Taipei CityTaiwan
| | - Chia‐Chao Wu
- Division of NephrologyDepartment of Internal MedicineTri‐Service General HospitalNational Defense Medical CenterTaipeiTaiwan
| | - Jia‐Feng Chang
- Department of Internal MedicineEn Chu Kong HospitalNew Taipei CityTaiwan
- Renal Care Joint FoundationNew Taipei CityTaiwan
| | - Jack L. Arbiser
- Department of DermatologyEmory School of Medicineand Winship Cancer InstituteAtlantaGAUSA
- Atlanta Veterans Administration Medical CenterDecaturGAUSA
| | - Chiz‐Tzung Chang
- Division of NephrologyDepartment of Internal MedicineChina Medical University HospitalTaichungTaiwan
| | - Ann Chen
- Graduate Institute of Life SciencesNational Defense Medical CenterTaipeiTaiwan
- Department of PathologyTri‐Service General HospitalNational Defense Medical CenterTaipeiTaiwan
| | - Shuk‐Man Ka
- Graduate Institute of Aerospace and Undersea MedicineDepartment of MedicineNational Defense Medical CenterTaipeiTaiwan
| |
Collapse
|
54
|
Gal R, Deres L, Horvath O, Eros K, Sandor B, Urban P, Soos S, Marton Z, Sumegi B, Toth K, Habon T, Halmosi R. Resveratrol Improves Heart Function by Moderating Inflammatory Processes in Patients with Systolic Heart Failure. Antioxidants (Basel) 2020; 9:E1108. [PMID: 33187089 PMCID: PMC7696241 DOI: 10.3390/antiox9111108] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022] Open
Abstract
The effects of resveratrol (RES) in heart failure have already been evaluated in animal models; however, in human clinical trials, they have not been confirmed yet. The aim of this study was to assess the effects of resveratrol treatment in systolic heart failure patients (heart failure with reduced ejection fraction or HFrEF). In this human clinical trial, 60 outpatients with NYHA (New York Heart Association) class II-III HFrEF were enrolled and randomized into two groups: receiving either 100-mg resveratrol daily or placebo for three months. At the beginning and at the end of the study echocardiography, a six-minute walk test, spirometry, quality of life questionnaire, lab test and RNA profile analysis were performed. The systolic and diastolic left ventricular function, as well as the global longitudinal strain, were improved significantly in the resveratrol-treated group (RES). Exercise capacity, ventilation parameters and quality of life also improved significantly in the RES group. In parallel, the cardiac biomarker levels (N-terminal prohormone of brain natriuretic peptide (NT-proBNP) and galectin-3) decreased in the treated group. The level of inflammatory cytokines decreased significantly after RES supplementation, as a consequence of the decreased expression level of leucocyte electron transport chain proteins. The main findings of our trial are that RES treatment added to the standard heart failure therapy improved heart function and the clinical condition by moderating the inflammatory processes in patients with HFrEF.
Collapse
Affiliation(s)
- Roland Gal
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7602 Pecs, Hungary; (R.G.); (L.D.); (O.H.); (B.S.); (Z.M.); (K.T.); (T.H.)
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
| | - Laszlo Deres
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7602 Pecs, Hungary; (R.G.); (L.D.); (O.H.); (B.S.); (Z.M.); (K.T.); (T.H.)
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1007 Budapest, Hungary
| | - Orsolya Horvath
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7602 Pecs, Hungary; (R.G.); (L.D.); (O.H.); (B.S.); (Z.M.); (K.T.); (T.H.)
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
| | - Krisztian Eros
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1007 Budapest, Hungary
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, 7602 Pecs, Hungary
| | - Barbara Sandor
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7602 Pecs, Hungary; (R.G.); (L.D.); (O.H.); (B.S.); (Z.M.); (K.T.); (T.H.)
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
| | - Peter Urban
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
| | - Szilvia Soos
- Division of Pulmonology, 1st Department of Medicine, Medical School, University of Pecs, 7602 Pecs, Hungary;
| | - Zsolt Marton
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7602 Pecs, Hungary; (R.G.); (L.D.); (O.H.); (B.S.); (Z.M.); (K.T.); (T.H.)
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
| | - Balazs Sumegi
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1007 Budapest, Hungary
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pecs, 7602 Pecs, Hungary
| | - Kalman Toth
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7602 Pecs, Hungary; (R.G.); (L.D.); (O.H.); (B.S.); (Z.M.); (K.T.); (T.H.)
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
| | - Tamas Habon
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7602 Pecs, Hungary; (R.G.); (L.D.); (O.H.); (B.S.); (Z.M.); (K.T.); (T.H.)
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
| | - Robert Halmosi
- Division of Cardiology, 1st Department of Medicine, Medical School, University of Pecs, 7602 Pecs, Hungary; (R.G.); (L.D.); (O.H.); (B.S.); (Z.M.); (K.T.); (T.H.)
- Szentágothai Research Centre, University of Pecs, 7602 Pecs, Hungary; (K.E.); (P.U.); (B.S.)
| |
Collapse
|
55
|
Hosseini A, Sheikh S, Soukhtanloo M, Malaekeh-Nikouei B, Rajabian A. The Effect of Hydro-alcoholic Extract of Rheum Turkestanicum Roots against Oxidative Stress in Endothelial Cells. Int J Prev Med 2020; 11:122. [PMID: 33088450 PMCID: PMC7554444 DOI: 10.4103/ijpvm.ijpvm_386_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 04/22/2020] [Indexed: 11/29/2022] Open
Abstract
Introduction: Cardiovascular disorders (CVD) are a common cause of mortality worldwide. Oxidative stress is thought to be a major factor leading to CVD. Anti-oxidants such as medicinal plants may have a role in the mitigation of vascular problems through free radicals scavenging. In this study, we evaluated the protective effects of Rheum turkestanicum against hydrogen peroxide (H2O2)-induced toxicity in endothelial cells (BAE-1). Methods: To evaluate the protective effect of R. turkestanicum against H2O2 toxicity, four groups comprised of control group (the cells without any treatment), H2O2 group (the cells incubated with H2O2 (200 μM)), and treatment groups (the cells treated with R. turkestanicum (12200 μg/ml) alone or 24h before exposure to H2O2). Quercetin (30.23 μg/ml) was used as a bioactive ingredient of the extract. Then the cell viability, reactive oxygen species, lipid peroxidation, and apoptosis were evaluated. Results: H2O2 exposure reduced cell viability to 13.6 ± 1.6%, enhanced ROS generation to 1445 ± 80.7%, lipid peroxidation (LPO, 290 ± 13% of control), and apoptotic cells (P < 0.001). In contrast, compared with H2O2 group, R. turkestanicum and quercetin significantly restored the cell viability to 80.3 ± 1.6 and 87.2 ± 2.1%, ROS formation to 186 ± 10 and 129 ± 1%, as well as LPO to 130.7 ± 7.7 and 116 ± 2.5 of control, respectively (P < 0.001). Therefore, the extract reduced H2O2-induced toxicity in BAE-1 cells by scavenging of free radicals. Conclusion: Our findings demonstrated that the extract might reduce toxicity of endothelial cells by attenuation of oxidative stress, which can be related to the presence of active ingredients including quercetin.
Collapse
Affiliation(s)
- Azar Hosseini
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Sheikh
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Medical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bizhan Malaekeh-Nikouei
- Nanotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Rajabian
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
56
|
Liu C, Ma X, Zhuang J, Liu L, Sun C. Cardiotoxicity of doxorubicin-based cancer treatment: What is the protective cognition that phytochemicals provide us? Pharmacol Res 2020; 160:105062. [DOI: 10.1016/j.phrs.2020.105062] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022]
|
57
|
Yang SR, Hsu WH, Wu CY, Shang HS, Liu FC, Chen A, Hua KF, Ka SM. Accelerated, severe lupus nephritis benefits from treatment with honokiol by immunoregulation and differentially regulating NF-κB/NLRP3 inflammasome and sirtuin 1/autophagy axis. FASEB J 2020; 34:13284-13299. [PMID: 32813287 DOI: 10.1096/fj.202001326r] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/08/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022]
Abstract
Using honokiol (HNK), a major anti-inflammatory bioactive compound in Magnolia officinalis, we show a potent therapeutic outcome against an accelerated, severe form of lupus nephritis (ASLN). The latter may follow infectious insults that act as environmental triggers in the patients. In the current study, an ASLN model in NZB/W F1 mice was treated with HNK by daily gavage after onset of the disease. We show that HNK ameliorated the ASLN by improving renal function, albuminuria, and renal pathology, especially reducing cellular crescents, neutrophil influx, fibrinoid necrosis in glomeruli, and glomerulonephritis activity scores. Meanwhile, HNK differentially regulated T cell functions, reduced serum anti-dsDNA autoantibodies, and inhibited NLRP3 inflammasome activation in the mice. The latter involved: (a) suppressed production of reactive oxygen species and NF-κB activation-mediated priming signal of the inflammasome, (b) reduced mitochondrial damage, and (c) enhanced sirtuin 1 (SIRT1)/autophagy axis activation. In conclusion, HNK represents a new drug candidate for acute, severe episodes of LN capable of alleviating renal lesions in ASLN mice by negatively regulating T cell functions and by enhancing SIRT1/autophagy axis-lessened NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Shin-Ruen Yang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wan-Han Hsu
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chung-Yao Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Sheng Shang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Feng-Cheng Liu
- Division of Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ann Chen
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Shuk-Man Ka
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Aerospace and Undersea Medicine, Department of Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
58
|
Long G, Chen H, Wu M, Li Y, Gao L, Huang S, Zhang Y, Jia Z, Xia W. Antianemia Drug Roxadustat (FG-4592) Protects Against Doxorubicin-Induced Cardiotoxicity by Targeting Antiapoptotic and Antioxidative Pathways. Front Pharmacol 2020; 11:1191. [PMID: 32848792 PMCID: PMC7419679 DOI: 10.3389/fphar.2020.01191] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/22/2020] [Indexed: 12/25/2022] Open
Abstract
Doxorubicin (DOX) is broadly used in treating various malignant tumors. However, its cardiotoxicity limits its clinical use. Roxadustat (FG-4592) is a new hypoxia-inducible factor prolyl hydroxylase (HIF-PHD) inhibitor and has been approved for treating anemia in chronic kidney diseases (CKD) patients. However, the role of FG-4592 in DOX-induced cardiotoxicity remains unknown. In this study, mouse cardiac function was evaluated by echocardiography, plasma LDH/CK-MB, and heart HE staining. Cell viability, apoptosis, oxidative stress, inflammation, and HIF-target genes were evaluated in mouse cardiac tissue and cardiac cells exposed to DOX with FG-4592 pretreatment. DOX-sensitive HepG2 and MCF-7 cell lines were used to evaluate FG-4592 effect on the anticancer activity of DOX. We found that FG-4592 alleviated DOX-induced cardiotoxicity shown by the protection against cardiac dysfunction, cardiac apoptosis, and oxidative stress without the effect on inflammatory response. FG-4592 alone did not change the cardiac function, cardiomyocyte morphology, oxidative stress, and inflammation in vivo. FG-4592 could protect cardiomyocytes against DOX-induced apoptosis and ROS production in line with the upregulation of HIF-1α and its target genes of Bcl-2 and SOD2. Importantly, FG-4592 displayed anticancer property in cancer cells treated with or without DOX. These findings highlighted the protective effect of FG-4592 on DOX-induced cardiotoxicity possibly through upregulating HIF-1α and its target genes antagonizing apoptosis and oxidative stress.
Collapse
Affiliation(s)
- Guangfeng Long
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Hongbing Chen
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Mengying Wu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yuanyuan Li
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Ling Gao
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Weiwei Xia
- Department of Clinical Laboratory, Children's Hospital of Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.,Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
59
|
Wu CY, Hua KF, Hsu WH, Suzuki Y, Chu LJ, Lee YC, Takahata A, Lee SL, Wu CC, Nikolic-Paterson DJ, Ka SM, Chen A. IgA Nephropathy Benefits from Compound K Treatment by Inhibiting NF-κB/NLRP3 Inflammasome and Enhancing Autophagy and SIRT1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:202-212. [PMID: 32482710 DOI: 10.4049/jimmunol.1900284] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/24/2020] [Indexed: 12/15/2022]
Abstract
IgA nephropathy (IgAN), the most common primary glomerular disorder, has a relatively poor prognosis yet lacks a pathogenesis-based treatment. Compound K (CK) is a major absorbable intestinal bacterial metabolite of ginsenosides, which are bioactive components of ginseng. The present study revealed promising therapeutic effects of CK in two complementary IgAN models: a passively induced one developed by repeated injections of IgA immune complexes and a spontaneously occurring model of spontaneous grouped ddY mice. The potential mechanism for CK includes 1) inhibiting the activation of NLRP3 inflammasome in renal tissues, macrophages and bone marrow-derived dendritic cells, 2) enhancing the induction of autophagy through increased SIRT1 expression, and 3) eliciting autophagy-mediated NLRP3 inflammasome inhibition. The results support CK as a drug candidate for IgAN.
Collapse
Affiliation(s)
- Chung-Yao Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan
| | - Wan-Han Hsu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan
| | - Lichieh Julie Chu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan 333, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital at Linkou, Gueishan, Taoyuan 333, Taiwan
| | - Yu-Chieh Lee
- Department of Biotechnology and Animal Science, National Ilan University, Ilan 260, Taiwan
| | - Akiko Takahata
- Department of Nephrology, Juntendo University Faculty of Medicine, Tokyo 113-8421, Japan
| | - Sheau-Long Lee
- Department of Chemistry, R.O.C. Military Academy, Kaohsiung 830, Taiwan
| | - Chia-Chao Wu
- Division of Nephrology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - David J Nikolic-Paterson
- Department of Nephrology and Monash University Centre for Inflammatory Diseases, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | - Shuk-Man Ka
- Graduate Institute of Aerospace and Undersea Medicine, Department of Medicine, National Defense Medical Center, Taipei 114, Taiwan; and
| | - Ann Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan;
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
60
|
Lou H, Yao J, Sun Y, Sun H, Song Z, Li H, Wang X, Liu K, Liu X, Li Z. Role of Blueberry Anthocyanin Extract in the Expression of SIRT1 and NF-κB in Rat Lens Epithelial Cells in Experimentally Induced DM. Curr Eye Res 2020; 46:45-51. [PMID: 32478572 DOI: 10.1080/02713683.2020.1776879] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE To investigate the mechanism of the protective effects of blueberry anthocyanin extract (BAE) against oxidative stress and the roles of SIRT1 and NF-κB in the pathogenesis of diabetic cataracts. METHODS Male SD rats were randomly divided into a control group (group A) and an experimental group. The rats in the experimental group were intraperitoneally injected with streptozotocin (STZ) (60 mg/kg). Rats with blood glucose levels ≥16.7 mmol/L were considered to have DM. The rats in the experimental group were subdivided into group B (distilled water by oral gavage: 10 ml/kg/day), group C (5% blueberry anthocyanin extract by oral gavage: 10 ml/kg/day), and group D (15% blueberry anthocyanin extract by oral gavage: 10 ml/kg/day), with 15 rats in each group. At the end of 8 weeks, some biochemical parameters, including the expression of SIRT1 and NF-κB by qRT-PCR and western blotting and the activity of SOD and GSH, were measured in lens epithelial cells (LECs). RESULTS The lenses of the rats in the control group appeared transparent during the entire 8-week period. Four weeks following STZ injection, cataracts gradually progressed in the experimental rats. SIRT1 expression was upregulated in groups B, C and D compared to the control group. However, the expression of NF-κB decreased in the experimental groups with increasing doses of BAE (p < .05). Our study also showed that the activity of the SOD enzyme and GSH in the LECs of the rats in the experimental group increased with higher doses of BAE. CONCLUSIONS The results indicated that BAE significantly delayed the progression of diabetic cataracts in rats. These effects may be due to the dose-dependent antioxidant activity of BAE, which is mediated by enhanced SOD and GSH activities, SIRT1 expression and reduced NF-κB expression. Abbreviations: SD rat: Sprague-Dawley rat; BAE: Blueberry anthocyanin extract; LECs: Lens epithelial cells; SOD: Superoxide dismutase; GSH: Glutathione; DM: Diabetes mellitus; SIRT1: Silent information regulator protein-1; STZ: Streptozotocin; PBS: Phosphate-buffered saline.
Collapse
Affiliation(s)
- Hong Lou
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China.,Department of Ophthalmology, Ningbo Eye Hospital , Ningbo, China
| | - Jiayu Yao
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China.,Department of Ophthalmology, Heihe People's Hospital , Heihe, China
| | - Ying Sun
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China.,Department of Ophthalmology, Second Hospital of Heilongjiang Province , Harbin, China
| | - Hong Sun
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China.,Department of Ophthalmology, Harbin 242 Hospital , Harbin, China
| | - Zhaowei Song
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China
| | - Huazhang Li
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China
| | - Xiaohui Wang
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China
| | - Kexin Liu
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China
| | - Xiangyu Liu
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China
| | - Zhijian Li
- Department of Ophthalmology, First Affiliated Hospital, Harbin Medical University , Harbin, China
| |
Collapse
|
61
|
Asnani A, Shi X, Farrell L, Lall R, Sebag IA, Plana JC, Gerszten RE, Scherrer-Crosbie M. Changes in Citric Acid Cycle and Nucleoside Metabolism Are Associated with Anthracycline Cardiotoxicity in Patients with Breast Cancer. J Cardiovasc Transl Res 2020; 13:349-356. [PMID: 31278494 DOI: 10.1007/s12265-019-09897-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 06/18/2019] [Indexed: 01/04/2023]
Abstract
Anthracyclines and HER2-targeted antibodies are very effective for the treatment of breast cancer, but their use is limited by cardiotoxicity. In this nested case-control study, we assessed the role of intermediary metabolism in 38 women with breast cancer treated with anthracyclines and trastuzumab. Using targeted mass spectrometry to measure 71 metabolites in the plasma, we identified changes in citric acid and aconitic acid that differentiated patients who developed cardiotoxicity from those who did not. In patients with cardiotoxicity, the magnitude of change in citric acid at three months correlated with the change in left ventricular ejection fraction (LVEF) and absolute LVEF at nine months. Patients with cardiotoxicity also demonstrated more pronounced changes in purine and pyrimidine metabolism. Early metabolic changes may therefore provide insight into the mechanisms associated with the development of chemotherapy-associated cardiotoxicity.
Collapse
Affiliation(s)
- Aarti Asnani
- CardioVascular Institute, Center for Life Sciences, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, 9th Floor, Boston, MA, 02115, USA.
- Cardiovascular Research Center and Corrigan Minehan Heart Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Xu Shi
- CardioVascular Institute, Center for Life Sciences, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, 9th Floor, Boston, MA, 02115, USA
- Cardiovascular Research Center and Corrigan Minehan Heart Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laurie Farrell
- CardioVascular Institute, Center for Life Sciences, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, 9th Floor, Boston, MA, 02115, USA
- Cardiovascular Research Center and Corrigan Minehan Heart Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Rahul Lall
- CardioVascular Institute, Center for Life Sciences, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, 9th Floor, Boston, MA, 02115, USA
| | - Igal A Sebag
- Sir Mortimer B. Davis-Jewish General Hospital and McGill University, Montreal, CA, USA
| | | | - Robert E Gerszten
- CardioVascular Institute, Center for Life Sciences, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, 9th Floor, Boston, MA, 02115, USA
- Cardiovascular Research Center and Corrigan Minehan Heart Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Marielle Scherrer-Crosbie
- CardioVascular Institute, Center for Life Sciences, Beth Israel Deaconess Medical Center and Harvard Medical School, 3 Blackfan Circle, 9th Floor, Boston, MA, 02115, USA
- Cardiovascular Research Center and Corrigan Minehan Heart Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
62
|
Patra S, Mishra SR, Behera BP, Mahapatra KK, Panigrahi DP, Bhol CS, Praharaj PP, Sethi G, Patra SK, Bhutia SK. Autophagy-modulating phytochemicals in cancer therapeutics: Current evidences and future perspectives. Semin Cancer Biol 2020; 80:205-217. [PMID: 32450139 DOI: 10.1016/j.semcancer.2020.05.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 05/11/2020] [Accepted: 05/13/2020] [Indexed: 02/08/2023]
Abstract
Autophagy is an intracellular catabolic self-cannibalism that eliminates dysfunctional cytoplasmic cargos by the fusion of cargo-containing autophagosomes with lysosomes to maintain cyto-homeostasis. Autophagy sustains a dynamic interlink between cytoprotective and cytostatic function during malignant transformation in a context-dependent manner. The antioxidant and immunomodulatory phyto-products govern autophagy and autophagy-associated signaling pathways to combat cellular incompetence during malignant transformation. Moreover, in a close cellular signaling circuit, autophagy regulates aberrant epigenetic modulation and inflammation, which limits tumor metastasis. Thus, manipulating autophagy for induction of cell death and associated regulatory phenomena will embark on a new strategy for tumor suppression with wide therapeutic implications. Despite the prodigious availability of lead pharmacophores in nature, the central autophagy regulating entities, their explicit target, as well as pre-clinical and clinical assessment remains a major question to be answered. In addition to this, the stage-specific regulation of autophagy and mode of action with natural products in regulating the key autophagic molecules, control of tumor-specific pathways in relation to modulation of autophagic network specify therapeutic target in caner. Moreover, the molecular pathway specificity and enhanced efficacy of the pre-existing chemotherapeutic agents in co-treatment with these phytochemicals hold high prevalence for target specific cancer therapeutics. Hence, the multi-specific role of phytochemicals in a cellular and tumor context dependent manner raises immense curiosity for investigating of novel therapeutic avenues. In this perspective, this review discusses about diverse implicit mechanisms deployed by the bioactive compounds in diagnosis and therapeutics approach during cancer progression with special insight into autophagic regulation.
Collapse
Affiliation(s)
- Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Soumya R Mishra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Bishnu P Behera
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Kewal K Mahapatra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Debasna P Panigrahi
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Chandra S Bhol
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Prakash P Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Samir K Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India
| | - Sujit K Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela 769008, Odisha, India.
| |
Collapse
|
63
|
The roles of resveratrol on cardiac mitochondrial function in cardiac diseases. Eur J Nutr 2020; 60:29-44. [PMID: 32372266 DOI: 10.1007/s00394-020-02256-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/22/2020] [Indexed: 12/31/2022]
Abstract
Left ventricular (LV) dysfunction is commonly associated with a variety of health conditions including acute myocardial infarction and obesity/diabetes. In addition, administration of several pharmacological agents such as anticancer, antiviral, and immunosuppressive drugs has been shown to be related with LV dysfunction. The molecular mechanism responsible for LV dysfunction has been extensively studied, and it has been proposed that the overproduction of reactive oxygen species (ROS) plays a crucial role in the regulation of this function. Mitochondria require the balance between ROS production and antioxidants to maintain their appropriate function and to prevent excessive ROS production. Thus, the excessive production of ROS and the reduced scavenging process under any pathological conditions could disrupt mitochondrial function, leading to energy depletion with subsequent cell death. Therefore, maintenance of the balance between oxidative stress and antioxidants is essential. Resveratrol, a stilbene, has been investigated extensively, and potentially used to treat or prevent various cardiovascular diseases. Resveratrol directly upregulates antioxidative capacity by increasing antioxidant genes such as heme oxygenase-1, superoxide dismutase, catalase, and glutathione. In this review, accumulated data from in vitro, ex vivo, and in vivo studies regarding the effects of resveratrol on cardiac mitochondrial function in cardiac pathologies are comprehensively summarized and discussed. Since there is no conclusive available clinical study regarding the effects of resveratrol on cardiac mitochondrial function, this review also aims to encourage more clinical investigations to confirm findings from basic research. This comprehensive review will provide insight regarding the potential mechanistic roles of resveratrol in preventing and/or treating patients with cardiovascular diseases to improve LV function and their health status.
Collapse
|
64
|
Tian W, Yang L, Liu Y, He J, Yang L, Zhang Q, Liu F, Li J, Liu J, Sumi S, Shen Y, Qi Z. Resveratrol attenuates doxorubicin-induced cardiotoxicity in rats by up-regulation of vascular endothelial growth factor B. J Nutr Biochem 2020; 79:108132. [DOI: 10.1016/j.jnutbio.2019.01.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/15/2019] [Accepted: 01/29/2019] [Indexed: 12/16/2022]
|
65
|
Chen J, Zhang S, Pan G, Lin L, Liu D, Liu Z, Mei S, Zhang L, Hu Z, Chen J, Luo H, Wang Y, Xin Y, You Z. Modulatory effect of metformin on cardiotoxicity induced by doxorubicin via the MAPK and AMPK pathways. Life Sci 2020; 249:117498. [PMID: 32142765 DOI: 10.1016/j.lfs.2020.117498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 03/02/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022]
Abstract
AIMS Doxorubicin (DOX) is an effective anthracycline anticancer drug. However, the clinical usage of it is limited due to its severe cardiotoxicity side effects. Metformin (Met) is a kind of first-line antihyperglycemic drug which has a potential protective effect on the heart,it is often used for oral treatment of type 2 diabetes. In this study, we explored whether Met could attenuate cardiotoxicity induced by DOX. MATERIALS AND METHODS For the sake of exploring the Met protective effect and mechanism, we established the DOX-induced cardiotoxicity models both in H9C2 cells incubated with 5 μM DOX in vitro and Sprague-Dawley rats treated with 20 mg/kg cumulative dose of DOX. KEY FINDINGS Met is able to inhibit growth inhibition and apoptosis of H9C2 cells induced by DOX. The heart indexes of rats were examined to evaluate the Met cardiotoxicity protection. Met improved the abnormal indexes, serum markers of cardiac heart injury, echocardiography, electrocardiogram, cardiac pathology, cardiomyocyte apoptosis, and oxidative stress markers induced by DOX. Furthermore, in vivo and in vitro studies demonstrated that Met protected against DOX-induced increasing cleaved caspase-3 and Bax. Met also prevented the downregulation of Bcl-2, activated the AMPK pathway, and inhibited the MAPK pathway. SIGNIFICANCE Met showed protective effects on DOX-induced cardiotoxicity by reducing oxidative stress and apoptosis, as well as regulating AMPK and MAPK signaling pathways.
Collapse
Affiliation(s)
- Jiaoting Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, China; Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Sheng Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Guixuan Pan
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lin Lin
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dongying Liu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhen Liu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Song Mei
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lijing Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhihang Hu
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jianguo Chen
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huaxing Luo
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yin Wang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Yanfei Xin
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Zhenqiang You
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
66
|
Sun Z, Lu W, Lin N, Lin H, Zhang J, Ni T, Meng L, Zhang C, Guo H. Dihydromyricetin alleviates doxorubicin-induced cardiotoxicity by inhibiting NLRP3 inflammasome through activation of SIRT1. Biochem Pharmacol 2020; 175:113888. [PMID: 32112883 DOI: 10.1016/j.bcp.2020.113888] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/24/2020] [Indexed: 12/21/2022]
Abstract
Doxorubicin (DOX) is a powerful anthracycline antineoplastic drug whose clinical application is limited by serious cardiotoxic side effects. Dihydromyricetin (DHM), a flavonoid compound extracted from the Japanese raisin tree (Hovenia dulcis), is cardioprotective in patients with heart failure; however, the underlying mechanisms are poorly understood. The aim of this study was to assess the possible anti-inflammatory properties of DHM in a rat model of DOX-induced cardiotoxicity and DOX-treated H9C2 cells, and gain insights into the molecular mechanisms that mediate these effects. The results showed that DHM treatment significantly improved the myocardial structure and function in DOX-exposed rats by alleviating NLRP3 inflammasome-mediated inflammation. DHM also inhibited DOX-induced activation of the NLRP3 inflammasome in H9C2 cells. This effect was mediated by inhibition of caspase-1 activity, suppression of IL-1β and IL-18 release, and upregulation of SIRT1 protein levels in vivo and in vitro. Moreover, selective inhibition of SIRT1 blocked the protective effects of DHM. Collectively, our findings indicate that DHM protects against DOX-induced cardiotoxicity by inhibiting NLRP3 inflammasome activation via stimulation of the SIRT1 pathway.
Collapse
Affiliation(s)
- Zhenzhu Sun
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Wenqiang Lu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Na Lin
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Jie Zhang
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Tingjuan Ni
- Zhejiang University School of Medicine, Hangzhou, China
| | - Liping Meng
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | | | - Hangyuan Guo
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China.
| |
Collapse
|
67
|
Resveratrol Prevents Right Ventricle Remodeling and Dysfunction in Monocrotaline-Induced Pulmonary Arterial Hypertension with a Limited Improvement in the Lung Vasculature. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1841527. [PMID: 32089765 PMCID: PMC7023844 DOI: 10.1155/2020/1841527] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/24/2019] [Accepted: 01/07/2020] [Indexed: 12/18/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a life-threatening disease that is characterized by an increase in pulmonary vascular pressure, leading to ventricular failure and high morbidity and mortality. Resveratrol, a phenolic compound and a sirtuin 1 pathway activator, has known dietary benefits and is used as a treatment for anti-inflammatory and cardiovascular diseases. Its therapeutic effects have been published in the scientific literature; however, its benefits in PAH are yet to be precisely elucidated. Using a murine model of PAH induced by monocrotaline, the macroscopic and microscopic effects of a daily oral dose of resveratrol in rats with PAH were evaluated by determining its impact on the lungs and the right and left ventricular function. While most literature has focused on smooth muscle cell mechanisms and lung pathology, our results highlight the relevance of therapy-mediated improvement of right ventricle and isolated cardiomyocyte physiology in both ventricles. Although significant differences in the pulmonary architecture were not identified either micro- or macroscopically, the effects of resveratrol on right ventricular function and remodeling were observed to be beneficial. The values for the volume, diameter, and contractility of the right ventricular cardiomyocytes returned to those of the control group, suggesting that resveratrol has a protective effect against ventricular dysfunction and pathological remodeling changes in PAH. The effect of resveratrol in the right ventricle delayed the progression of findings associated with right heart failure and had a limited positive effect on the architecture of the lungs. The use of resveratrol could be considered a future potential adjunct therapy, especially when the challenges to making a diagnosis and the current therapy limitations for PAH are taken into consideration.
Collapse
|
68
|
Pervaiz S, Bellot GL, Lemoine A, Brenner C. Redox signaling in the pathogenesis of human disease and the regulatory role of autophagy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 352:189-214. [DOI: 10.1016/bs.ircmb.2020.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
69
|
Sarina, Li DF, Feng ZQ, Du J, Zhao WH, Huang N, Jia JC, Wu ZY, Alamusi, Wang YY, Ji XL, Yu L. Mechanism of Placenta Damage in Gestational Diabetes Mellitus by Investigating TXNIP of Patient Samples and Gene Functional Research in Cell Line. Diabetes Ther 2019; 10:2265-2288. [PMID: 31654346 PMCID: PMC6848504 DOI: 10.1007/s13300-019-00713-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM) is a gestational complication that affects maternal and child health. The placenta provides the fetus with the necessary nutrition and oxygen and takes away the metabolic waste. Patients with GDM are diagnosed and treated merely on the basis of the blood glucose level; this approach does nothing to help evaluate the status of the placenta, which is worth noting in GDM. The purpose of this research was to clarify the relation between thioredoxin-interacting protein (TXNIP) and reactive oxygen species (ROS) in the placenta of patients with GDM, which has thus far remained unclear. METHODS The expression of TXNIP in the placentas of 10 patients with GDM and 10 healthy puerperae (control group) was investigated via immunofluorescence. The relation among TXNIP, ROS, and the function of mitochondria was explored in HTR-8/SVneo cells stimulated by high glucose (HG). RESULTS The results showed the expression of TXNIP in the placentas of patients with GDM was higher than that in the control group, and the expression of TXNIP in HTR-8/SVneo cells treated with HG was higher than that in the control group, causing the accumulation of ROS and changes of mitochondria, promoting apoptosis and inhibition of migration. CONCLUSIONS High expression of TXNIP caused by HG mediates the increasing ROS and the mitochondria dysfunction in GDM; this impairs the function of the placenta and is the basis for the prediction of perinatal outcome.
Collapse
Affiliation(s)
- Sarina
- Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Dong Fang Li
- Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Zong Qi Feng
- Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Jie Du
- Department of Gynecology and Obstetrics, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Wen Hua Zhao
- Department of Gynecology and Obstetrics, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Na Huang
- Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Jian Chao Jia
- Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Zhou Ying Wu
- Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Alamusi
- Department of Ophthalmology, Inner Mongolia International Mongolian Hospital, Hohhot, 010000, China
| | - Yong Yun Wang
- Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Xiao Li Ji
- Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot, 010010, China
| | - Lan Yu
- Department of Clinical Medical Research Center, Inner Mongolia People's Hospital, Hohhot, 010010, China.
| |
Collapse
|
70
|
Ferreira LL, Cervantes M, Froufe HJC, Egas C, Cunha-Oliveira T, Sassone-Corsi P, Oliveira PJ. Doxorubicin persistently rewires cardiac circadian homeostasis in mice. Arch Toxicol 2019; 94:257-271. [PMID: 31768571 DOI: 10.1007/s00204-019-02626-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/13/2019] [Indexed: 12/29/2022]
Abstract
Circadian rhythms disruption can be the cause of chronic diseases. External cues, including therapeutic drugs, have been shown to modulate peripheral-circadian clocks. Since anthracycline cardiotoxicity is associated with loss of mitochondrial function and metabolic remodeling, we investigated whether the energetic failure induced by sub-chronic doxorubicin (DOX) treatment in juvenile mice was associated with persistent disruption of circadian regulators. Juvenile C57BL/6J male mice were subjected to a sub-chronic DOX treatment (4 weekly injections of 5 mg/kg DOX) and several cardiac parameters, as well as circadian-gene expression and acetylation patterns, were analyzed after 6 weeks of recovery time. Complementary experiments were performed with Mouse Embryonic Fibroblasts (MEFs) and Human Embryonic Kidney 293 cells. DOX-treated juvenile mice showed cardiotoxicity markers and persistent alterations of transcriptional- and signaling cardiac circadian homeostasis. The results showed a delayed influence of DOX on gene expression, accompanied by changes in SIRT1-mediated cyclic deacetylation. The mechanism behind DOX interference with the circadian clock was further studied in vitro, in which were observed alterations of circadian-gene expression and increased BMAL1 SIRT1-mediated deacetylation. In conclusion, DOX treatment in juvenile mice resulted in disruption of oscillatory molecular mechanisms including gene expression and acetylation profiles.
Collapse
Affiliation(s)
- Luciana L Ferreira
- Mitochondrial Toxicology and Experimental Therapeutics Laboratory (MitoXT), CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building (Lote 8A), Biocant Park, 3060-197, Cantanhede, Portugal
| | - Marlene Cervantes
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, University of California, Irvine, CA, 92697, USA
| | - Hugo J C Froufe
- Next Generation Sequencing Unit, Biocant, Biocant Park, Núcleo 04, Lote 8, Cantanhede, Portugal
| | - Conceição Egas
- Mitochondrial Toxicology and Experimental Therapeutics Laboratory (MitoXT), CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building (Lote 8A), Biocant Park, 3060-197, Cantanhede, Portugal.,Next Generation Sequencing Unit, Biocant, Biocant Park, Núcleo 04, Lote 8, Cantanhede, Portugal
| | - Teresa Cunha-Oliveira
- Mitochondrial Toxicology and Experimental Therapeutics Laboratory (MitoXT), CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building (Lote 8A), Biocant Park, 3060-197, Cantanhede, Portugal
| | - Paolo Sassone-Corsi
- Department of Biological Chemistry, Center for Epigenetics and Metabolism, University of California, Irvine, CA, 92697, USA
| | - Paulo J Oliveira
- Mitochondrial Toxicology and Experimental Therapeutics Laboratory (MitoXT), CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building (Lote 8A), Biocant Park, 3060-197, Cantanhede, Portugal. .,Institute for Interdisciplinary Research (I.I.I.), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
71
|
Liu D, Ma Z, Xu L, Zhang X, Qiao S, Yuan J. PGC1α activation by pterostilbene ameliorates acute doxorubicin cardiotoxicity by reducing oxidative stress via enhancing AMPK and SIRT1 cascades. Aging (Albany NY) 2019; 11:10061-10073. [PMID: 31733141 PMCID: PMC6914429 DOI: 10.18632/aging.102418] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/28/2019] [Indexed: 01/23/2023]
Abstract
Doxorubicin (DOX) is a widely used and potent anticancer agent, but DOX dose-dependently induced cardiotoxicity greatly limits its use in clinic. Pterostilbene, a natural analog of resveratrol, is a known antioxidant and exerts myocardial protection. The present study explored the action and detailed mechanism of pterostilbene on DOX-treated cardiomyocytes. We investigated the effects of pterostilbene on established acute DOX-induced cardiotoxicity models in both H9c2 cells treated with 1 μM DOX and C57BL/6 mice with DOX (20 mg/kg cumulative dose) exposure. Pterostilbene markedly alleviated the DOX exposure-induced acute myocardial injury. Both in vitro and in vivo studies revealed that pterostilbene inhibited the acute DOX exposure-caused oxidative stress and mitochondrial morphological disorder via the PGC1α upregulation through activating AMPK and via PGC1α deacetylation through enhancing SIRT1. However, these effects were partially reversed by knockdown of AMPK or SIRT1 in vitro and treatment of Compound C (AMPK inhibitor) or EX527 (SIRT1 inhibitor) in vivo. Our results indicate that pterostilbene protects cardiomyocytes from acute DOX exposure-induced oxidative stress and mitochondrial damage via PGC1α upregulation and deacetylation through activating AMPK and SIRT1 cascades.
Collapse
Affiliation(s)
- Dong Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an 710038, China
| | - Liqun Xu
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an 710038, China
| | - Xiaoyan Zhang
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an 710038, China
| | - Shubin Qiao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Jiansong Yuan
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| |
Collapse
|
72
|
The anti-cancer drug doxorubicin induces substantial epigenetic changes in cultured cardiomyocytes. Chem Biol Interact 2019; 313:108834. [PMID: 31545955 DOI: 10.1016/j.cbi.2019.108834] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023]
Abstract
The anthracycline doxorubicin (DOX) is widely used in cancer therapy with the limitation of cardiotoxicity leading to the development of congestive heart failure. DOX-induced oxidative stress and changes of the phosphoproteome as well as epigenome were described but the exact mechanisms of the adverse long-term effects are still elusive. Here, we tested the impact of DOX treatment on cell death, oxidative stress parameters and expression profiles of proteins involved in epigenetic pathways in a cardiomyocyte cell culture model. Markers of oxidative stress, apoptosis and expression of proteins involved in epigenetic processes were assessed by immunoblotting in cultured rat myoblasts (H9c2) upon treatment with DOX (1 or 5 μM for 24 or 48 h) in adherent viable and detached apoptotic cells. The apoptosis markers cleaved caspase-3 and fractin as well as oxidative stress markers 3-nitrotyrosine and malondialdehyde were dose-dependently increased by DOX treatment. Histone deacetylases (SIRT1 and HDAC2), histone lysine demethylases (KDM3A and LSD1) and histone lysine methyltransferases (SET7 and SMYD1) were significantly regulated by DOX treatment with generation of cleaved protein fragments and posttranslational modifications. Overall, we found significant decrease in histone 3 acetylation in DOX-treated cells. DOX treatment of cultured cardiomyocyte precursor cells causes severe cell death by apoptosis associated with cellular oxidative stress. In addition, significant regulation of proteins involved in epigenetic processes and changes in global histone 3 acetylation were observed. However, the significance and clinical impact of these changes remain elusive.
Collapse
|
73
|
Song S, Chu L, Liang H, Chen J, Liang J, Huang Z, Zhang B, Chen X. Protective Effects of Dioscin Against Doxorubicin-Induced Hepatotoxicity Via Regulation of Sirt1/FOXO1/NF-κb Signal. Front Pharmacol 2019; 10:1030. [PMID: 31572199 PMCID: PMC6753638 DOI: 10.3389/fphar.2019.01030] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (Dox), an antitumor antibiotic, has therapeutic effects on many kinds of tumors. However, Dox can produce some serious side effects that limit its clinical application. Thus, exploration of effective drug targets or active lead compounds against Dox-induced organ damage is necessary. Dioscin, one natural product, has potent effects against Dox-induced renal injury and cardiotoxicity. However, the effects of dioscin on Dox-induced hepatotoxicity have not been reported. In this study, the results showed that dioscin significantly ameliorated Dox-induced cell injury, reduced reactive oxygen species (ROS) level, and suppressed cell apoptosis in alpha mouse liver 12 (AML-12) cells caused by Dox. In vivo, dioscin evidently decreased the levels of alanine transaminase (ALT), aspartate transaminase (AST), malondialdehyde (MDA); increased the levels of superoxide dismutase (SOD), glutathione (GSH), and glutathione peroxidase (GSH-Px); and alleviated liver injury. Mechanism study showed that dioscin remarkably up-regulated the expression levels of silent information regulator 1 (Sirt1) and heme oxygenase-1 (HO-1) via increase of the nuclear translocation of NF-E2-related factor 2 (Nrf2) and suppressed the expression levels of forkhead box protein O1 (FOXO1) and kelch-like ECH-associated protein-1 (Keap1) to inhibit oxidative stress. Furthermore, dioscin obviously decreased the nuclear translocation of nuclear factor κB (NF-κB) and the mRNA levels of tumor necrosis factor alpha (TNF-α), interleukin 1β (IL-1β), and interleukin 6 (IL-6) to suppress inflammation. Meanwhile, dioscin significantly regulated tumor suppressor P53 (P53) expression level and BCL-2-associated X (BAX)/BCL-2 apoptosis regulator (BCL-2) ratio to inhibit cell apoptosis. These results were further validated by knockdown of Sirt1 using siRNA silencing in AML-12 cells, which confirmed that the target of dioscin against Dox-induced hepatotoxicity was Sirt1/FOXO1/NF-κB signal. In short, our findings showed that dioscin exhibited protective effects against Dox-induced liver damage via suppression of oxidative stress, inflammation, and apoptosis, which should be developed as one new candidate for the prevention of Dox-induced liver injury in the future.
Collapse
Affiliation(s)
- Shasha Song
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Jin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| |
Collapse
|
74
|
Tan X, Li L, Wang J, Zhao B, Pan J, Wang L, Liu X, Liu X, Liu Z. Resveratrol Prevents Acrylamide-Induced Mitochondrial Dysfunction and Inflammatory Responses via Targeting Circadian Regulator Bmal1 and Cry1 in Hepatocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8510-8519. [PMID: 31294559 DOI: 10.1021/acs.jafc.9b03368] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Acrylamide, mainly formed in Maillard browning reaction during food processing, causes defects in liver circadian clock and mitochondrial function by inducing oxidative stress. Resveratrol is a polyphenol that has powerful antioxidant and anti-inflammatory activity. However, the preventive effects of resveratrol on acrylamide-triggered oxidative damage and circadian rhythm disorders are unclear at the current stage. The present research revealed that resveratrol pretreatment prevented acrylamide-induced cell death, mitochondrial dysfunction, and inflammatory responses in HepG2 liver cells. Acrylamide significantly triggered disorders of circadian genes transcription and protein expressions including Bmal1 and Cry 1 in primary hepatocytes, which were prevented by resveratrol pretreatment. Moreover, we found that the beneficial effects of resveratrol on stimulating Nrf2/NQO-1 pathway and mitochondrial respiration complex expressions in acrylamide-treated cells were Bmal1-dependent. Similarly, the inhibitory effects of resveratrol on inflammation signaling NF-κB were Cry1-dependent. In conclusion, these results demonstrated resveratrol could be a promising compound in suppressing acrylamide-induced hepatotoxicity and balancing the circadian clock.
Collapse
Affiliation(s)
- Xintong Tan
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Yangling 712100 , China
| | - Ling Li
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Yangling 712100 , China
| | - Jia Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Yangling 712100 , China
| | - Beita Zhao
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Yangling 712100 , China
| | - Junru Pan
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Yangling 712100 , China
| | - Leran Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Yangling 712100 , China
| | - Xiao Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Yangling 712100 , China
| | - Xuebo Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Yangling 712100 , China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering , Northwest A&F University , Yangling 712100 , China
| |
Collapse
|
75
|
Yamasaki S, Tomihara T, Kimura G, Ueno Y, Ketema RM, Sato S, Mukai Y, Sikder T, Kurasaki M, Hosokawa T, Saito T. Long-term effects of maternal resveratrol intake during lactation on cholesterol metabolism in male rat offspring. Int J Food Sci Nutr 2019; 71:226-234. [PMID: 31290360 DOI: 10.1080/09637486.2019.1639638] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Resveratrol (RSV) can protect against non-communicable diseases by improving cholesterol metabolism. However, it is unclear that effects of maternal RSV intake on health of adult offspring. In this study, we examined effects of maternal RSV intake during lactation on cholesterol metabolism in adult male rat offspring. Female Wistar rats were fed a control diet (CON) supplemented with or without RSV (20 mg/kg body weight/day) during their lactation period. Male offspring were weaned onto a standard diet and maintained on this diet for 36 weeks. As a result, plasma cholesterol level significantly decreased in RSV offspring compared to CON offspring. Furthermore, a decrease in hepatic 3-hydroxy-3-methylglutaryl-CoA reductase level and an increase in hepatic LDL-receptor level were observed in the RSV offspring. These results indicate that maternal RSV intake causes long-term decrease in plasma cholesterol level in the offspring through suppression of hepatic cholesterol biosynthesis and promotion of hepatic cholesterol uptake.
Collapse
Affiliation(s)
- Shojiro Yamasaki
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Tomomi Tomihara
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Goh Kimura
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yukako Ueno
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan
| | | | - Shin Sato
- Department of Nutrition, Aomori University of Health and Welfare, Aomori, Japan
| | - Yuuka Mukai
- Faculty of Health and Social Work, Kanagawa University of Human Services, Yokosuka, Japan
| | - Tajuddin Sikder
- Department of Public Health and Informatics, Jahangirnagar University, Dhaka, Bangladesh
| | - Masaaki Kurasaki
- Faculty of Environmental Earth Science, Hokkaido University, Sapporo, Japan
| | - Toshiyuki Hosokawa
- Institute for the Advancement of Higher Education, Hokkaido University, Sapporo, Japan
| | - Takeshi Saito
- Graduate School of Health Sciences, Hokkaido University, Sapporo, Japan.,Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| |
Collapse
|
76
|
Abdelgawad IY, Grant MKO, Zordoky BN. Leveraging the Cardio-Protective and Anticancer Properties of Resveratrol in Cardio-Oncology. Nutrients 2019; 11:nu11030627. [PMID: 30875799 PMCID: PMC6471701 DOI: 10.3390/nu11030627] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 03/08/2019] [Accepted: 03/09/2019] [Indexed: 12/25/2022] Open
Abstract
Cardio-oncology is a clinical/scientific discipline which aims to prevent and/or treat cardiovascular diseases in cancer patients. Although a large number of cancer treatments are known to cause cardiovascular toxicity, they are still widely used because they are highly effective. Unfortunately, therapeutic interventions to prevent and/or treat cancer treatment-induced cardiovascular toxicity have not been established yet. A major challenge for such interventions is to protect the cardiovascular system without compromising the therapeutic benefit of anticancer medications. Intriguingly, the polyphenolic natural compound resveratrol and its analogs have been shown in preclinical studies to protect against cancer treatment-induced cardiovascular toxicity. They have also been shown to possess significant anticancer properties on their own, and to enhance the anticancer effect of other cancer treatments. Thus, they hold significant promise to protect the cardiovascular system and fight the cancer at the same time. In this review, we will discuss the current knowledge regarding the cardio-protective and the anticancer properties of resveratrol and its analogs. Thereafter, we will discuss the challenges that face the clinical application of these agents. To conclude, we will highlight important gaps of knowledge and future research directions to accelerate the translation of these exciting preclinical findings to cancer patient care.
Collapse
Affiliation(s)
- Ibrahim Y Abdelgawad
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Marianne K O Grant
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
77
|
Detection of anthracycline-induced cardiotoxicity using perfusion-corrected 99mTc sestamibi SPECT. Sci Rep 2019; 9:216. [PMID: 30659226 PMCID: PMC6338786 DOI: 10.1038/s41598-018-36721-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 11/23/2018] [Indexed: 01/02/2023] Open
Abstract
By the time cardiotoxicity-associated cardiac dysfunction is detectable by echocardiography it is often beyond meaningful intervention. 99mTc-sestamibi is used clinically to image cardiac perfusion by single photon emission computed tomography (SPECT) imaging, but as a lipophilic cation its distribution is also governed by mitochondrial membrane potential (ΔΨm). Correcting scans for variations in perfusion (using a ΔΨm-independent perfusion tracer such as (bis(N-ethoxy-N-ethyldithiocarbamato)nitrido 99mTc(V)) (99mTc-NOET) could allow 99mTc-sestamibi to be repurposed to specifically report on ΔΨm as a readout of evolving cardiotoxicity. Isolated rat hearts were perfused within a γ-detection apparatus to characterize the pharmacokinetics of 99mTc-sestamibi and 99mTc-NOET in response to mitochondrial perturbation by hypoxia, ionophore (CCCP) or doxorubicin. All interventions induced 99mTc-sestamibi washout; hypoxia from 24.9 ± 2.6% ID to 0.4 ± 6.2%, CCCP from 22.8 ± 2.5% ID to −3.5 ± 3.1%, and doxorubicin from 23.0 ± 2.2% ID to 17.8 ± 0.7, p < 0.05. Cardiac 99mTc-NOET retention (34.0 ± 8.0% ID) was unaffected in all cases. Translating to an in vivo rat model, 2 weeks after bolus doxorubicin injection, there was a dose-dependent loss of cardiac 99mTc-sestamibi retention (from 2.3 ± 0.3 to 0.9 ± 0.2 ID/g with 10 mg/kg (p < 0.05)), while 99mTc-NOET retention (0.93 ± 0.16 ID/g) was unaffected. 99mTc-NOET therefore traps in myocardium independently of the mitochondrial perturbations that induce 99mTc-sestamibi washout, demonstrating proof-of-concept for an imaging approach to detect evolving cardiotoxicity.
Collapse
|
78
|
Cao S, Shen Z, Wang C, Zhang Q, Hong Q, He Y, Hu C. Resveratrol improves intestinal barrier function, alleviates mitochondrial dysfunction and induces mitophagy in diquat challenged piglets1. Food Funct 2019; 10:344-354. [DOI: 10.1039/c8fo02091d] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study evaluated whether resveratrol can alleviate intestinal injury and enhance the mitochondrial function and the mitophagy level in diquat induced oxidative stress of piglets.
Collapse
Affiliation(s)
- Shuting Cao
- Animal Science College
- Zhejiang University
- Key Laboratory of Molecular Animal Nutrition
- Ministry of Education
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province
| | - Zhuojun Shen
- Animal Science College
- Zhejiang University
- Key Laboratory of Molecular Animal Nutrition
- Ministry of Education
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province
| | - Chunchun Wang
- Animal Science College
- Zhejiang University
- Key Laboratory of Molecular Animal Nutrition
- Ministry of Education
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province
| | - Qianhui Zhang
- Animal Science College
- Zhejiang University
- Key Laboratory of Molecular Animal Nutrition
- Ministry of Education
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province
| | - Qihua Hong
- Animal Science College
- Zhejiang University
- Key Laboratory of Molecular Animal Nutrition
- Ministry of Education
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province
| | - Yonghui He
- Henan Province Engineering Technology Centre of Intelligent Cleaner Production of Livestock and Poultry
- Henan Institute of Science and Technology
- Xinxiang
- China
| | - Caihong Hu
- Animal Science College
- Zhejiang University
- Key Laboratory of Molecular Animal Nutrition
- Ministry of Education
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province
| |
Collapse
|
79
|
Schlüter KD, Kutsche HS, Hirschhäuser C, Schreckenberg R, Schulz R. Review on Chamber-Specific Differences in Right and Left Heart Reactive Oxygen Species Handling. Front Physiol 2018; 9:1799. [PMID: 30618811 PMCID: PMC6304434 DOI: 10.3389/fphys.2018.01799] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/29/2018] [Indexed: 01/21/2023] Open
Abstract
Reactive oxygen species (ROS) exert signaling character (redox signaling), or damaging character (oxidative stress) on cardiac tissue depending on their concentration and/or reactivity. The steady state of ROS concentration is determined by the interplay between its production (mitochondrial, cytosolic, and sarcolemmal enzymes) and ROS defense enzymes (mitochondria, cytosol). Recent studies suggest that ROS regulation is different in the left and right ventricle of the heart, specifically by a different activity of superoxide dismutase (SOD). Mitochondrial ROS defense seems to be lower in right ventricular tissue compared to left ventricular tissue. In this review we summarize the current evidence for heart chamber specific differences in ROS regulation that may play a major role in an observed inability of the right ventricle to compensate for cardiac stress such as pulmonary hypertension. Based on the current knowledge regimes to increase ROS defense in right ventricular tissue should be in the focus for the development of future therapies concerning right heart failure.
Collapse
Affiliation(s)
| | - Hanna Sarah Kutsche
- Department of Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | | | - Rolf Schreckenberg
- Department of Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Rainer Schulz
- Department of Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
80
|
Benzer F, Kandemir FM, Kucukler S, Comaklı S, Caglayan C. Chemoprotective effects of curcumin on doxorubicin-induced nephrotoxicity in wistar rats: by modulating inflammatory cytokines, apoptosis, oxidative stress and oxidative DNA damage. Arch Physiol Biochem 2018; 124:448-457. [PMID: 29302997 DOI: 10.1080/13813455.2017.1422766] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Doxorubicin (DXR) is one of the most important chemotherapeutic agent. However, nephrotoxicity reduces its clinical utility in humans. The aim of the study was to investigate protective effects of curcumin (CMN) against DXR-induced nephrotoxicity. Rats were subjected to oral treatment of CMN (100 and 200 mg/kg body weight) for 7 days. Nephrotoxicity was induced by single intra peritoneal injection of DXR (40 mg/kg body weight) on the fifth day and then the experiment was terminated on the eighth day. Nephroprotective effects of CMN were associated with decrease in serum toxicity markers and increase in antioxidant enzyme activities. CMN was able to reduced the levels of inflammatory markers such as TNF-α, NF-κB, IL-1β, iNOS and COX-2 in the rats. It also reduced the expressions of apoptotic marker including caspase-3, and oxidative DNA damage marker including 8-OHdG. Collectively, these findings indicated that CMN protect against DXR-induced nephrotoxicity.
Collapse
Affiliation(s)
- Fulya Benzer
- a Department of Food Engineering, Faculty of Engineering , Munzur University , Tunceli , Turkey
| | - Fatih Mehmet Kandemir
- b Department of Biochemistry, Faculty of Veterinary Medicine , Ataturk University , Erzurum , Turkey
| | - Sefa Kucukler
- b Department of Biochemistry, Faculty of Veterinary Medicine , Ataturk University , Erzurum , Turkey
| | - Selim Comaklı
- c Department of Pathology, Faculty of Veterinary Medicine , Ataturk University , Erzurum , Turkey
| | - Cuneyt Caglayan
- d Department of Biochemistry, Faculty of Veterinary Medicine , Bingol University , Bingol , Turkey
| |
Collapse
|
81
|
Manna D, Bhuyan R, Ghosh R. Probing the mechanism of SIRT1 activation by a 1,4-dihydropyridine. J Mol Model 2018; 24:340. [PMID: 30448921 DOI: 10.1007/s00894-018-3877-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 11/05/2018] [Indexed: 11/25/2022]
Abstract
The NAD+-dependent deacetylase SIRT1 plays important roles in several physiological processes such as transcription, genome stability, stress responses, and aging. Due to its diverse role in metabolisms, SIRT1 has emerged as a potential therapeutic target in many human disorders such as type II diabetes, cardiovascular and neurodegenerative diseases, and cancer. Recent studies have reported that modulation of SIRT1 activity by phenolic activators like resveratrol and some 1,4-dihydropyridines (1,4-DHPs) can inhibit tumor growth by promoting apoptosis in cancer cells. However, the mechanism of SIRT1 activation is still not clear. In this report, we have tried to elucidate the mechanism of SIRT1 activation from studies on its interaction with a synthetic 1,4-DHP derivative (DHP-8; 3,5-diethoxy carbonyl-4-(4-nitrophenyl)-2,6-dimethyl-1,4-dihydropyridine) using molecular modeling, docking, simulation, and free energy analyses. Owing to the absence of full-length human SIRT1 structure, multi-template based modeling approach was opted followed by docking of DHP-8 at its allosteric site. In presence of DHP-8, the overall conformation of SIRT1 was found to be more stable (especially at its substrate binding sites) with a large structural variation at its N-terminal domain while bound to substrate p53 or p53-W. Determination of the MM/PBSA free energy indicated that the binding of DHP-8 to SIRT1 significantly increased the binding affinity of SIRT1 to its substrate p53-W as well as to NAD+. Overall, this study depicts the atomistic detailed mechanism for the direct activation of SIRT1 by a 1,4-DHP. This would serve to develop new SIRT1 activators for future therapeutic perspectives.
Collapse
Affiliation(s)
- Debashri Manna
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Rajabrata Bhuyan
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Rita Ghosh
- Department of Biochemistry and Biophysics, University of Kalyani, Nadia, West Bengal, 741235, India.
| |
Collapse
|
82
|
Zhang QL, Yang JJ, Zhang HS. Carvedilol (CAR) combined with carnosic acid (CAA) attenuates doxorubicin-induced cardiotoxicity by suppressing excessive oxidative stress, inflammation, apoptosis and autophagy. Biomed Pharmacother 2018; 109:71-83. [PMID: 30396094 DOI: 10.1016/j.biopha.2018.07.037] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 07/07/2018] [Accepted: 07/07/2018] [Indexed: 01/10/2023] Open
Abstract
Doxorubicin (DOX) is a wide spectrum antitumor drug. However, its clinical application is limited due to the cardiotoxicity. Carvedilol (CAR) is a β-blocker used to treat high blood pressure and heart failure. Accordingly, supplementation with natural antioxidants or plant extracts exerts protective effects against various injury in vivo. Carnosic acid (CAA), the principal constituent of rosemary, has various biological activities, including antioxidant, antitumor, and anti-inflammatory. Here, heart injury mouse model was established using DOX (20 mg/kg) in vivo. And cardiac muscle cell line of H9C2 was subjected to 0.5 μM of DOX for 24 h in vitro. Then, the protective effects of CAA and CAR alone, or the two in combination on DOX-induced cardiotoxicity in vivo and in vitro were explored. The results indicated that both CAA and CAR, when used alone, were moderately effective in attenuating DOX-induced cardiotoxicity. The combination of two drugs functioned synergistically to ameliorate cardiac injury caused by DOX, as evidenced by the significantly reduced collagen accumulation and improved dysfunction of heart. CAA and CAR exhibited stronger anti-oxidative role in DOX-treated mice partly by augmenting the expression and activities of the anti-oxidative enzymes. In addition, inflammatory response was significantly suppressed by the two in combination, proved by the decreased pro-inflammatory cytokines (COX2, TNF-α, IL-6, IL-1β and IL-18), which was associated with the inactivation of nuclear factor κB (NF-κB). Furthermore, DOX-stirred apoptosis and autophagy were dramatically attenuated by the co-treatments of CAA and CAR through down-regulating cleaved Caspase-3 and LC3B signaling pathways. The effects of CAA and CAR combination against cardiotoxicity were observed in H9C2 cells with DOX stimulation. Our findings above suggested that the use of CAR and CAA in combination could be expected to have synergistic efficacy and significant potential against cardiotoxicity induced by DOX.
Collapse
Affiliation(s)
- Qiu-Lan Zhang
- Department of Cardiology, Jining Second People's Hospital, Jining 272000, China
| | - Jing-Jie Yang
- Department of Emergency, Liaocheng People's Hospital, Liaocheng 252000, China
| | - Hong-Sheng Zhang
- Department of Cardiology, Affiliated Hospital of Jining Medical University, 272000, China.
| |
Collapse
|
83
|
Synthesis of a Reactive Oxygen Species-Responsive Doxorubicin Derivative. Molecules 2018; 23:molecules23071809. [PMID: 30037071 PMCID: PMC6100310 DOI: 10.3390/molecules23071809] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/16/2018] [Accepted: 07/18/2018] [Indexed: 01/01/2023] Open
Abstract
A heterobifunctional reactive oxygen species (ROS)-responsive linker for directed drug assembly onto and delivery from a quantum dot (QD) nanoparticle carrier was synthesized and coupled to doxorubicin using N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide hydrochloride (EDC)/sulfo–NHS coupling. The doxorubicin conjugate was characterized using 1H NMR and LC-MS and subsequently reacted under conditions of ROS formation (Cu2+/H2O2) resulting in successful and rapid thioacetal oxidative cleavage, which was monitored using 1H NMR.
Collapse
|
84
|
Govender J, Loos B, Marais E, Engelbrecht AM. Melatonin improves cardiac and mitochondrial function during doxorubicin-induced cardiotoxicity: A possible role for peroxisome proliferator-activated receptor gamma coactivator 1-alpha and sirtuin activity? Toxicol Appl Pharmacol 2018; 358:86-101. [PMID: 29966675 DOI: 10.1016/j.taap.2018.06.031] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/27/2018] [Accepted: 06/28/2018] [Indexed: 01/02/2023]
Abstract
Mitochondrial dysfunction is a central element in the development of doxorubicin (DXR)-induced cardiotoxicity. In this context, melatonin is known to influence mitochondrial homeostasis and function. This study aimed to investigate the effects of melatonin on cardiac function, tumor growth, mitochondrial fission and fusion, PGC1-α and sirtuin activity in an acute model of DXR-induced cardiotoxicity. During the in vitro study, H9c2 rat cardiomyoblasts were pre-treated with melatonin (10 μM, 24 h) followed by DXR exposure (3 μM, 24 h). Following treatment, cellular ATP levels and mitochondrial morphology were assessed. In the in vivo study, female Sprague Dawley rats (16 weeks old), were inoculated with a LA7 rat mammary tumor cell line and tumors were measure daily. Animals were injected with DXR (3 × 4 mg/kg) and/or received melatonin (6 mg/kg) for 14 days in their drinking water. Rat hearts were used to conduct isolated heart perfusions to assess cardiac function and thereafter, heart tissue was used for immunoblot analysis. DXR treatment increased cell death and mitochondrial fission which were reduced with melatonin treatment. Cardiac output increased in rats treated with DXR + melatonin compared to DXR-treated rats. Tumor volumes was significantly reduced in DXR + melatonin-treated rats on Day 8 in comparison to DXR-treated rats. Furthermore, DXR + melatonin treatment increased cellular ATP levels, PGC1-α and SIRT1 expression which was attenuated by DXR treatment. These results indicate that melatonin treatment confers a dual cardio-protective and oncostatic effect by improving mitochondrial function and cardiac function whilst simultaneously retarding tumor growth during DXR-induced cardiotoxicity.
Collapse
Affiliation(s)
- Jenelle Govender
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa.
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Erna Marais
- Department of Medical Physiology, Faculty of Medicine, Stellenbosch University, Tygerberg Campus, 7505, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
85
|
Giorgi C, Marchi S, Simoes IC, Ren Z, Morciano G, Perrone M, Patalas-Krawczyk P, Borchard S, Jȩdrak P, Pierzynowska K, Szymański J, Wang DQ, Portincasa P, Wȩgrzyn G, Zischka H, Dobrzyn P, Bonora M, Duszynski J, Rimessi A, Karkucinska-Wieckowska A, Dobrzyn A, Szabadkai G, Zavan B, Oliveira PJ, Sardao VA, Pinton P, Wieckowski MR. Mitochondria and Reactive Oxygen Species in Aging and Age-Related Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 340:209-344. [PMID: 30072092 PMCID: PMC8127332 DOI: 10.1016/bs.ircmb.2018.05.006] [Citation(s) in RCA: 248] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aging has been linked to several degenerative processes that, through the accumulation of molecular and cellular damage, can progressively lead to cell dysfunction and organ failure. Human aging is linked with a higher risk for individuals to develop cancer, neurodegenerative, cardiovascular, and metabolic disorders. The understanding of the molecular basis of aging and associated diseases has been one major challenge of scientific research over the last decades. Mitochondria, the center of oxidative metabolism and principal site of reactive oxygen species (ROS) production, are crucial both in health and in pathogenesis of many diseases. Redox signaling is important for the modulation of cell functions and several studies indicate a dual role for ROS in cell physiology. In fact, high concentrations of ROS are pathogenic and can cause severe damage to cell and organelle membranes, DNA, and proteins. On the other hand, moderate amounts of ROS are essential for the maintenance of several biological processes, including gene expression. In this review, we provide an update regarding the key roles of ROS-mitochondria cross talk in different fundamental physiological or pathological situations accompanying aging and highlighting that mitochondrial ROS may be a decisive target in clinical practice.
Collapse
Affiliation(s)
- Carlotta Giorgi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Saverio Marchi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Ines C.M. Simoes
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ziyu Ren
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Giampaolo Morciano
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
- Maria Pia Hospital, GVM Care & Research, Torino, Italy
| | - Mariasole Perrone
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paulina Patalas-Krawczyk
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Sabine Borchard
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Paulina Jȩdrak
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | | | - Jȩdrzej Szymański
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - David Q. Wang
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Dept. of Biomedical Sciences & Human Oncology, University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Grzegorz Wȩgrzyn
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, Munich, Germany
| | - Pawel Dobrzyn
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Massimo Bonora
- Departments of Cell Biology and Gottesman Institute for Stem Cell & Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jerzy Duszynski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Alessandro Rimessi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | | | | | - Gyorgy Szabadkai
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Barbara Zavan
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Paulo J. Oliveira
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, Biocant Park, University of Coimbra, Cantanhede, Portugal
| | - Vilma A. Sardao
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, Biocant Park, University of Coimbra, Cantanhede, Portugal
| | - Paolo Pinton
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
| | - Mariusz R. Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
86
|
Shi W, Deng H, Zhang J, Zhang Y, Zhang X, Cui G. Mitochondria-Targeting Small Molecules Effectively Prevent Cardiotoxicity Induced by Doxorubicin. Molecules 2018; 23:E1486. [PMID: 29921817 PMCID: PMC6099719 DOI: 10.3390/molecules23061486] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/13/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
Doxorubicin (Dox) is a chemotherapeutic agent widely used for the treatment of numerous cancers. However, the clinical use of Dox is limited by its unwanted cardiotoxicity. Mitochondrial dysfunction has been associated with Dox-induced cardiotoxicity. To mitigate Dox-related cardiotoxicity, considerable successful examples of a variety of small molecules that target mitochondria to modulate Dox-induced cardiotoxicity have appeared in recent years. Here, we review the related literatures and discuss the evidence showing that mitochondria-targeting small molecules are promising cardioprotective agents against Dox-induced cardiac events.
Collapse
Affiliation(s)
- Wei Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| | - Hongkuan Deng
- School of Life Sciences, Shandong University of Technology, Zibo 255000, China.
| | - Jianyong Zhang
- Pharmacy School, Zunyi Medical University, Zunyi 563003, China.
| | - Ying Zhang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| | - Xiufang Zhang
- School of Life Sciences, Shandong University of Technology, Zibo 255000, China.
| | - Guozhen Cui
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China.
| |
Collapse
|
87
|
Resveratrol Ameliorates Microcystin-LR-Induced Testis Germ Cell Apoptosis in Rats via SIRT1 Signaling Pathway Activation. Toxins (Basel) 2018; 10:toxins10060235. [PMID: 29890735 PMCID: PMC6024601 DOI: 10.3390/toxins10060235] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/03/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022] Open
Abstract
Microcystin-leucine arginine (MC-LR), a cyclic heptapeptide produced by cyanobacteria, is a strong reproductive toxin. Studies performed in rat Sertoli cells and Chinese hamster ovary cells have demonstrated typical apoptosis after MC-LR exposure. However, little is known on how to protect against the reproductive toxicity induced by MC-LR. The present study aimed to explore the possible molecular mechanism underlying the anti-apoptosis and protective effects of resveratrol (RES) on the co-culture of Sertoli–germ cells and rat testes. The results demonstrated that MC-LR treatment inhibited the proliferation of Sertoli–germ cells and induced apoptosis. Furthermore, sirtuin 1 (SIRT1) and Bcl-2 were inhibited, while p53 and Ku70 acetylation, Bax expression, and cleaved caspase-3 were upregulated by MC-LR. However, RES pretreatment ameliorated MC-LR-induced apoptosis and SIRT1 inhibition, and downregulated the MC-LR-induced increase in p53 and Ku70 acetylation, Bax expression, and caspase-3 activation. In addition, RES reversed the MC-LR-mediated reduction in Ku70 binding to Bax. The present study indicated that the administration of RES could ameliorate MC-LR-induced Sertoli–germ cell apoptosis and protect against reproductive toxicity in rats by stimulating the SIRT1/p53 pathway, suppressing p53 and Ku70 acetylation and enhancing the binding of Ku70 to Bax.
Collapse
|
88
|
Yang X, Liu N, Li X, Yang Y, Wang X, Li L, Jiang L, Gao Y, Tang H, Tang Y, Xing Y, Shang H. A Review on the Effect of Traditional Chinese Medicine Against Anthracycline-Induced Cardiac Toxicity. Front Pharmacol 2018; 9:444. [PMID: 29867456 PMCID: PMC5963334 DOI: 10.3389/fphar.2018.00444] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 04/16/2018] [Indexed: 01/29/2023] Open
Abstract
Anthracyclines are effective agents generally used to treat solid-tumor and hematologic malignancies. The use of anthracyclines for over 40 years has improved cancer survival statistics. Nevertheless, the clinical utility of anthracyclines is limited by its dose-dependent cardiotoxicity that adversely affects 10-30% of patients. Anthracycline-induced cardiotoxicity may be classified as acute/subacute or chronic/late toxicity and leads to devastating adverse effects resulting in poor quality of life, morbidity, and premature mortality. Traditional Chinese medicine has a history of over 2,000 years, involving both unique theories and substantial experience. Several studies have investigated the potential of natural products to decrease the cardiotoxic effects of chemotherapeutic agents on healthy cells, without negatively affecting their antineoplastic activity. This article discusses the mechanism of anthracycline-induced cardiotoxicity, and summarizes traditional Chinese medicine treatment for anthracycline-induced heart failure (HF), cardiac arrhythmia, cardiomyopathy, and myocardial ischemia in recent years, in order to provide a reference for the clinical prevention and treatment of cardiac toxicity.
Collapse
Affiliation(s)
- Xinyu Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Nian Liu
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Xinye Li
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yihan Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaofeng Wang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Linling Li
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Le Jiang
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Hebin Tang
- Department of Pharmacology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yong Tang
- Department of Pancreatic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yanwei Xing
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
89
|
Burns KE, Delehanty JB. Cellular delivery of doxorubicin mediated by disulfide reduction of a peptide-dendrimer bioconjugate. Int J Pharm 2018; 545:64-73. [PMID: 29709616 DOI: 10.1016/j.ijpharm.2018.04.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/08/2018] [Accepted: 04/13/2018] [Indexed: 02/07/2023]
Abstract
In this study, we developed a peptide-dendrimer-drug conjugate system for the pH-triggered direct cytosolic delivery of the cancer chemotherapeutic doxorubicin (DOX) using the pH Low Insertion Peptide (pHLIP). We synthesized a pHLIP-dendrimer-DOX conjugate in which a single copy of pHLIP displayed a generation three dendrimer bearing multiple copies of DOX via disulfide linkages. Biophysical analysis showed that both the dendrimer and a single DOX conjugate inserted into membrane bilayers in a pH-dependent manner. Time-resolved confocal microscopy indicate the single DOX conjugate may undergo a faster rate of membrane translocation, due to greater nuclear localization of DOX at 24 h and 48 h post delivery. At 72 h, however, the levels of DOX nuclear accumulation for both constructs were identical. Cytotoxicity assays revealed that both constructs mediated ∼80% inhibition of cellular proliferation at 10 µM, the dendrimer complex exhibited a 17% greater cytotoxic effect at lower concentrations and greater than three-fold improvement in IC50 over free DOX. Our findings show proof of concept that the dendrimeric display of DOX on the pHLIP carrier (1) facilitates the pH-dependent and temporally-controlled release of DOX to the cytosol, (2) eliminates the endosomal sequestration of the drug cargo, and (3) augments DOX cytotoxicity relative to the free drug.
Collapse
Affiliation(s)
- Kelly E Burns
- Center for Bio/Molecular Science and Engineering, U.S. Naval Research Laboratory, Code 6900, Washington DC 20375, United States; National Research Council, Washington DC 20001, United States
| | - James B Delehanty
- Center for Bio/Molecular Science and Engineering, U.S. Naval Research Laboratory, Code 6900, Washington DC 20375, United States.
| |
Collapse
|
90
|
Cucurbitacin I Protects H9c2 Cardiomyoblasts against H 2O 2-Induced Oxidative Stress via Protection of Mitochondrial Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3016382. [PMID: 29682157 PMCID: PMC5845511 DOI: 10.1155/2018/3016382] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/08/2018] [Accepted: 01/31/2018] [Indexed: 12/25/2022]
Abstract
Cucurbitacin I, a triterpenoid natural compound, exhibits various pharmacological properties, including anticancer, anti-inflammatory, and hepatoprotective properties. However, antioxidant effects of cucurbitacin I in cardiac cells are currently unknown. In the present study, we assessed the preventive effects of cucurbitacin I against the oxidative stress in H9c2 cardiomyoblasts. To evaluate antioxidant effects of cucurbitacin I in H9c2 cardiomyoblasts, H2O2-treated H9c2 cells were pretreated with various concentrations of the cucurbitacin I. Cell viability, reactive oxygen species (ROS) production, and apoptosis were determined to elucidate the protective effects of cucurbitacin I against H2O2-induced oxidative stress in H9c2 cells. In addition, we assessed the mitochondrial functions and protein expression levels of mitogen-activated protein kinases (MAPKs). Cucurbitacin I prevented the cells against cell death and ROS production and elevated the antioxidant protein levels upon oxidative stress. Furthermore, cucurbitacin I preserved the mitochondrial functions and inhibited the apoptotic responses in H2O2-treated cells. Cucurbitacin I also suppressed the activation of MAPK proteins (extracellular signal-regulated kinase 1/2, c-Jun N-terminal kinase, and p38). Collectively, cucurbitacin I potentially protects the H9c2 cardiomyoblasts against oxidative stress and further suggests that it can be utilized as a therapeutic agent for the prevention of oxidative stress in cardiac injury.
Collapse
|
91
|
Yu J, Wang C, Kong Q, Wu X, Lu JJ, Chen X. Recent progress in doxorubicin-induced cardiotoxicity and protective potential of natural products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 40:125-139. [PMID: 29496165 DOI: 10.1016/j.phymed.2018.01.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 12/26/2017] [Accepted: 01/14/2018] [Indexed: 05/24/2023]
Abstract
BACKGROUND As an anthracycline antibiotic, doxorubicin (DOX) is one of the most potent and widely used chemotherapeutic agents for various types of solid tumors. Unfortunately, clinical application of this drug results in severe side effects of cardiotoxicity. PURPOSE We aim to review the research focused on elimination or reduction of DOX cardiotoxicity without affecting its anticancer efficacy by natural products. METHODS This study is based on pertinent papers that were retrieved by a selective search using relevant keywords in PubMed and ScienceDirect. The literature mainly focusing on natural products and herb extracts with therapeutic efficacies against experimental models both in vitro and in vivo was identified. RESULTS Current evidence revealed that multiple molecules and signaling pathways, such as oxidative stress, iron metabolism, and inflammation, are associated with DOX-induced cardiotoxicity. Based on these knowledge, various strategies were proposed, and thousands of compounds were screened. A number of natural products and herb extracts demonstrated potency in limiting DOX cardiotoxicity toward cultured cells and experimental animal models. CONCLUSIONS Though a panel of natural products and herb extracts demonstrate protective effects on DOX-induced cardiotoxicity in cells and animal models, their therapeutic potentials for clinical needs further investigation.
Collapse
Affiliation(s)
- Jie Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China
| | - Changxi Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China
| | - Qi Kong
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing 100021, PR China
| | - Xiaxia Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macau, PR China.
| |
Collapse
|
92
|
Sheena TS, Balaji P, Venkatesan R, Akbarsha MA, Jeganathan K. Functional evaluation of doxorubicin decorated polymeric liposomal curcumin: a surface tailored therapeutic platform for combination chemotherapy. NEW J CHEM 2018. [DOI: 10.1039/c8nj02406e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The present study deals with the hypothesis and design of new platform for the accommodation of curcumin and doxorubicin in surface engineered liposomes for combination chemotherapy.
Collapse
Affiliation(s)
- Thankaraj Salammal Sheena
- Centre for Nanoscience and Nanotechnology
- Department of Physics
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| | - Perumalsamy Balaji
- National Centre for Alternatives to Animal Experiments (NCAAE)
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| | - Rajiu Venkatesan
- Centre for Nanoscience and Nanotechnology
- Department of Physics
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| | | | - K. Jeganathan
- Centre for Nanoscience and Nanotechnology
- Department of Physics
- Bharathidasan University
- Tiruchirappalli – 620 024
- India
| |
Collapse
|
93
|
Sangtani A, Petryayeva E, Wu M, Susumu K, Oh E, Huston AL, Lasarte-Aragones G, Medintz IL, Algar WR, Delehanty JB. Intracellularly Actuated Quantum Dot–Peptide–Doxorubicin Nanobioconjugates for Controlled Drug Delivery via the Endocytic Pathway. Bioconjug Chem 2017; 29:136-148. [DOI: 10.1021/acs.bioconjchem.7b00658] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Ajmeeta Sangtani
- Fischell
Department of Bioengineering, University of Maryland, College Park, Maryland 20742, United States
| | - Eleonora Petryayeva
- Department
of Chemistry, University of British Columbia, Vancouver, British Columbia Canada V6T 1Z1
| | - Miao Wu
- Department
of Chemistry, University of British Columbia, Vancouver, British Columbia Canada V6T 1Z1
| | | | - Eunkeu Oh
- KeyW Corporation, Hanover, Maryland 21076, United States
| | | | | | | | - W. Russ Algar
- Department
of Chemistry, University of British Columbia, Vancouver, British Columbia Canada V6T 1Z1
| | | |
Collapse
|
94
|
Fernandes GFS, Silva GDB, Pavan AR, Chiba DE, Chin CM, Dos Santos JL. Epigenetic Regulatory Mechanisms Induced by Resveratrol. Nutrients 2017; 9:nu9111201. [PMID: 29104258 PMCID: PMC5707673 DOI: 10.3390/nu9111201] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022] Open
Abstract
Resveratrol (RVT) is one of the main natural compounds studied worldwide due to its potential therapeutic use in the treatment of many diseases, including cancer, diabetes, cardiovascular diseases, neurodegenerative diseases and metabolic disorders. Nevertheless, the mechanism of action of RVT in all of these conditions is not completely understood, as it can modify not only biochemical pathways but also epigenetic mechanisms. In this paper, we analyze the biological activities exhibited by RVT with a focus on the epigenetic mechanisms, especially those related to DNA methyltransferase (DNMT), histone deacetylase (HDAC) and lysine-specific demethylase-1 (LSD1).
Collapse
Affiliation(s)
- Guilherme Felipe Santos Fernandes
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
- Institute of Chemistry, São Paulo State University (UNESP), 14800060 Araraquara, Brazil.
| | | | - Aline Renata Pavan
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
| | - Diego Eidy Chiba
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
| | - Chung Man Chin
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
| | - Jean Leandro Dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), 14800903 Araraquara, Brazil.
| |
Collapse
|
95
|
Yuan YP, Ma ZG, Zhang X, Xu SC, Zeng XF, Yang Z, Deng W, Tang QZ. CTRP3 protected against doxorubicin-induced cardiac dysfunction, inflammation and cell death via activation of Sirt1. J Mol Cell Cardiol 2017; 114:38-47. [PMID: 29061338 DOI: 10.1016/j.yjmcc.2017.10.008] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 10/04/2017] [Accepted: 10/20/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Inflammation and myocytes apoptosis play critical roles in the development of doxorubicin (DOX)-induced cardiotoxicity. Our previous study found that C1q/tumour necrosis factor-related protein-3 (CTRP3) could inhibit cardiac inflammation and apoptosis of myocytes but its role in DOX-induced heart injury remains largely unknown. Our study aimed to investigate whether CTRP3 protected against DOX-induced heart injury and the underlying mechanism. METHODS We overexpressed CTRP3 in the hearts using an adeno-associated virus system. The mice were subjected to a single intraperitoneal injection of DOX (15mg/kg) to induce short-term model for cardiomyopathy. The morphological examination and biochemical analysis were used to evaluate the effects of CTRP3. H9C2 cells were used to verify the protective role of CTRP3 in vitro. RESULTS Myocardial CTRP3 protein levels were reduced in DOX-treated mice. Cardiac specific-overexpression of CTRP3 preserved heart dysfunction, and attenuated cardiac inflammation and cell loss induced by DOX in vivo and in vitro. CTRP3 could activate silent information regulator 1 (Sirt1) in vivo and in vitro. Moreover, specific inhibitor of Sirt1 and the silence of Sirt1 could abolish the protective effects of CTRP3 against DOX-induced inflammation and apoptosis. CONCLUSION CTRP3 protected against DOX-induced heart injury via activation of Sirt1. CTRP3 has therapeutic potential for the treatment of DOX cardiotoxicity.
Collapse
Affiliation(s)
- Yu-Pei Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Si-Chi Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Xiao-Feng Zeng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Cardiology, Wuhan 430060, PR China.
| |
Collapse
|
96
|
Resveratrol induces mitochondria-mediated, caspase-independent apoptosis in murine prostate cancer cells. Oncotarget 2017; 8:20895-20908. [PMID: 28157696 PMCID: PMC5400554 DOI: 10.18632/oncotarget.14947] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 01/10/2017] [Indexed: 12/29/2022] Open
Abstract
Found in the skins of red fruits, including grapes, resveratrol (RES) is a polyphenolic compound with cancer chemopreventive activity. Because of this activity, it has gained interest for scientific investigations. RES inhibits tumor growth and progression by targeting mitochondria-dependent or -independent pathways. However, further investigations are needed to explore the underlying mechanisms. The present study is focused on examining the role of RES-induced, mitochondria-mediated, caspase-independent apoptosis of prostate cancer cells, namely transgenic adenocarcinoma of mouse prostate (TRAMP) cells. These cells were exposed to RES for various times, and cell killing, cell morphology, mitochondrial membrane potential (Δψm), expression of Bax and Bcl2 proteins, the role of caspase-3, and DNA fragmentation were analyzed. TRAMP cells exposed to RES showed decreased cell viability, altered cell morphology, and disrupted Δψm, which led to aberrant expression of Bax and Bcl2 proteins. Furthermore, since the caspase-3 inhibitor, z-VAD-fmk (benzyloxycarbonyl-valine-alanine-aspartic acid-fluoromethyl ketone), had no appreciable impact on RES-induced cell killing, the killing was evidently caspase-independent. In addition, RES treatment of TRAMP-C1, TRAMP-C2, and TRAMP-C3 cells caused an appreciable breakage of genomic DNA into low-molecular-weight fragments. These findings show that, in inhibition of proliferation of TRAMP cells, RES induces mitochondria-mediated, caspase-independent apoptosis. Therefore, RES may be utilized as a therapeutic agent to control the proliferation and growth of cancer cells.
Collapse
|
97
|
Cardioprotective Effect of Resveratrol in a Postinfarction Heart Failure Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6819281. [PMID: 29109832 PMCID: PMC5646324 DOI: 10.1155/2017/6819281] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/06/2017] [Indexed: 01/25/2023]
Abstract
Despite great advances in therapies observed during the last decades, heart failure (HF) remained a major health problem in western countries. In order to further improve symptoms and survival in patients with heart failure, novel therapeutic strategies are needed. In some animal models of HF resveratrol (RES), it was able to prevent cardiac hypertrophy, contractile dysfunction, and remodeling. Several molecular mechanisms are thought to be involved in its protective effects, such as inhibition of prohypertrophic signaling molecules, improvement of myocardial Ca2+ handling, regulation of autophagy, and the reduction of oxidative stress and inflammation. In our present study, we wished to further examine the effects of RES on prosurvival (Akt-1, GSK-3β) and stress signaling (p38-MAPK, ERK 1/2, and MKP-1) pathways, on oxidative stress (iNOS, COX-2 activity, and ROS formation), and ultimately on left ventricular function, hypertrophy and fibrosis in a murine, and isoproterenol- (ISO-) induced postinfarction heart failure model. RES treatment improved left ventricle function, decreased interstitial fibrosis, cardiac hypertrophy, and the level of plasma BNP induced by ISO treatment. ISO also increased the activation of P38-MAPK, ERK1/2Thr183-Tyr185, COX-2, iNOS, and ROS formation and decreased the phosphorylation of Akt-1, GSK-3β, and MKP-1, which were favorably influenced by RES. According to our results, regulation of these pathways may also contribute to the beneficial effects of RES in HF.
Collapse
|
98
|
Altered mitochondrial epigenetics associated with subchronic doxorubicin cardiotoxicity. Toxicology 2017; 390:63-73. [PMID: 28865727 DOI: 10.1016/j.tox.2017.08.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 08/25/2017] [Accepted: 08/29/2017] [Indexed: 12/25/2022]
Abstract
Doxorubicin (DOX), a potent and broad-spectrum antineoplastic agent, causes an irreversible, cumulative and dose-dependent cardiomyopathy that ultimately leads to congestive heart failure. The mechanisms responsible for DOX cardiotoxicity remain poorly understood, but seem to involve mitochondrial dysfunction on several levels. Epigenetics may explain a portion of this effect. Since mitochondrial dysfunction may affect the epigenetic landscape, we hypothesize that this cardiac toxicity may result from epigenetic changes related to disruption of mitochondrial function. To test this hypothesis, eight-week-old male Wistar rats (n=6/group) were administered 7 weekly injections with DOX (2mgkg-1) or saline, and sacrificed two weeks after the last injection. We assessed gene expression patterns by qPCR, global DNA methylation by ELISA, and proteome lysine acetylation status by Western blot in cardiac tissue from saline and DOX-treated rats. We show for the first time that DOX treatment decreases global DNA methylation in heart but not in liver. These differences were accompanied by alterations in mRNA expression of multiple functional gene groups. DOX disrupted cardiac mitochondrial biogenesis, as demonstrated by decreased mtDNA levels and altered transcript levels for multiple mitochondrial genes encoded by both nuclear and mitochondrial genomes. Transcription of genes involved in lipid metabolism and epigenetic modulation were also affected. Western blotting analyses indicated a differential protein acetylation pattern in cardiac mitochondrial fractions of DOX-treated rats compared to controls. Additionally, DOX treatment increased the activity of histone deacetylases. These results suggest an interplay between mitochondrial dysfunction and epigenetic alterations, which may be a primary determinant of DOX-induced cardiotoxicity.
Collapse
|
99
|
Dolinsky VW. The role of sirtuins in mitochondrial function and doxorubicin-induced cardiac dysfunction. Biol Chem 2017; 398:955-974. [PMID: 28253192 DOI: 10.1515/hsz-2016-0316] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/11/2017] [Indexed: 01/15/2023]
Abstract
Anthracycline chemotherapeutics such as doxorubicin continue to be important treatments for many cancers. Through improved screening and therapy, more patients are surviving and living longer after the diagnosis of their cancer. However, anthracyclines are associated with both short- and long-term cardiotoxic effects. Doxorubicin-induced mitochondrial dysfunction is a central mechanism in the cardiotoxic effects of doxorubicin that contributes to impaired cardiac energy levels, increased reactive oxygen species production, cardiomyocyte apoptosis and the decline in cardiac function. Sirtuins are protein deacetylases that are activated by low energy levels and stimulate energy production through their activation of transcription factors and enzymatic regulators of cardiac energy metabolism. In addition, sirtuins activate oxidative stress resistance pathways. SIRT1 and SIRT3 are expressed at high levels in the cardiomyocyte. This review examines the function of sirtuins in the regulation of cardiac mitochondrial function, with a focus on their role in heart failure and an emphasis on their effects on doxorubicin-induced cardiotoxicity. We discuss the potential for sirtuin activation in combination with anthracycline chemotherapy in order to mitigate its cardiotoxic side-effects without reducing the antineoplastic activity of anthracyclines.
Collapse
|
100
|
Mattera R, Benvenuto M, Giganti MG, Tresoldi I, Pluchinotta FR, Bergante S, Tettamanti G, Masuelli L, Manzari V, Modesti A, Bei R. Effects of Polyphenols on Oxidative Stress-Mediated Injury in Cardiomyocytes. Nutrients 2017; 9:nu9050523. [PMID: 28531112 PMCID: PMC5452253 DOI: 10.3390/nu9050523] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/09/2017] [Accepted: 05/16/2017] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases are the main cause of mortality and morbidity in the world. Hypertension, ischemia/reperfusion, diabetes and anti-cancer drugs contribute to heart failure through oxidative and nitrosative stresses which cause cardiomyocytes nuclear and mitochondrial DNA damage, denaturation of intracellular proteins, lipid peroxidation and inflammation. Oxidative or nitrosative stress-mediated injury lead to cardiomyocytes apoptosis or necrosis. The reactive oxygen (ROS) and nitrogen species (RNS) concentration is dependent on their production and on the expression and activity of anti-oxidant enzymes. Polyphenols are a large group of natural compounds ubiquitously expressed in plants, and epidemiological studies have shown associations between a diet rich in polyphenols and the prevention of various ROS-mediated human diseases. Polyphenols reduce cardiomyocytes damage, necrosis, apoptosis, infarct size and improve cardiac function by decreasing oxidative stress-induced production of ROS or RNS. These effects are achieved by the ability of polyphenols to modulate the expression and activity of anti-oxidant enzymes and several signaling pathways involved in cells survival. This report reviews current knowledge on the potential anti-oxidative effects of polyphenols to control the cardiotoxicity induced by ROS and RNS stress.
Collapse
Affiliation(s)
- Rosanna Mattera
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Maria Gabriella Giganti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Ilaria Tresoldi
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | | | - Sonia Bergante
- IRCCS "S. Donato" Hospital, San Donato Milanese, Piazza Edmondo Malan, 20097 Milan, Italy.
| | - Guido Tettamanti
- IRCCS "S. Donato" Hospital, San Donato Milanese, Piazza Edmondo Malan, 20097 Milan, Italy.
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome "Sapienza", 00164 Rome, Italy.
| | - Vittorio Manzari
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Andrea Modesti
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
- Center for Regenerative Medicine (CIMER), University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy.
- Center for Regenerative Medicine (CIMER), University of Rome "Tor Vergata", 00133 Rome, Italy.
| |
Collapse
|