1
|
Cai Z, Xu S, Liu C. Cathepsin B in cardiovascular disease: Underlying mechanisms and therapeutic strategies. J Cell Mol Med 2024; 28:e70064. [PMID: 39248527 PMCID: PMC11382359 DOI: 10.1111/jcmm.70064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 09/10/2024] Open
Abstract
Cathepsin B (CTSB) is a member of the cysteine protease family, primarily responsible for degrading unnecessary organelles and proteins within the acidic milieu of lysosomes to facilitate recycling. Recent research has revealed that CTSB plays a multifaceted role beyond its function as a proteolytic enzyme in lysosomes. Importantly, recent data suggest that CTSB has significant impacts on different cardiac pathological conditions, such as atherosclerosis (AS), myocardial infarction, hypertension, heart failure and cardiomyopathy. Especially in the context of AS, preclinical models and clinical sample imaging data indicate that the cathepsin activity-based probe can reliably image CTSB activity in foam cells and atherosclerotic plaques; concurrently, it allows synchronous diagnostic and therapeutic interventions. However, our knowledge of CTSB in cardiovascular disease is still in the early stage. This paper aims to provide a comprehensive review of the significance of CTSB in cardiovascular physiology and pathology, with the objective of laying a theoretical groundwork for the development of drugs targeting CTSB.
Collapse
Affiliation(s)
- Zhulan Cai
- Department of Cardiology, Peking University Third Hospital, Beijing, P.R. China
| | - Shunyao Xu
- Department of Critical Care Medicine, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, P.R. China
| | - Chen Liu
- Department of Geriatrics, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, P.R. China
| |
Collapse
|
2
|
Ebbert L, von Montfort C, Wenzel CK, Reichert AS, Stahl W, Brenneisen P. A Combination of Cardamonin and Doxorubicin Selectively Affect Cell Viability of Melanoma Cells: An In Vitro Study. Antioxidants (Basel) 2024; 13:864. [PMID: 39061932 PMCID: PMC11274308 DOI: 10.3390/antiox13070864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Treatment of the most aggressive and deadliest form of skin cancer, the malignant melanoma, still has room for improvement. Its invasive nature and ability to rapidly metastasize and to develop resistance to standard treatment often result in a poor prognosis. While the highly effective standard chemotherapeutic agent doxorubicin (DOX) is widely used in a variety of cancers, systemic side effects still limit therapy. Especially, DOX-induced cardiotoxicity remains a big challenge. In contrast, the natural chalcone cardamonin (CD) has been shown to selectively kill tumor cells. Besides its anti-tumor activity, CD exhibits anti-oxidative, anti-inflammatory and anti-bacterial properties. In this study, we investigated the effect of the combinational treatment of DOX with CD on A375 melanoma cells compared to normal human dermal fibroblasts (NHDF) and rat cardiac myoblasts (H9C2 cells). DOX-induced cytotoxicity was unselective and affected all cell types, especially H9C2 cardiac myoblasts, demonstrating its cardiotoxic effect. In contrast, CD only decreased the cell viability of A375 melanoma cells, without harming normal (healthy) cells. The addition of CD selectively protected human dermal fibroblasts and rat cardiac myoblasts from DOX-induced cytotoxicity. While no apoptosis was induced by the combinational treatment in normal (healthy) cells, an apoptosis-mediated cytotoxicity was demonstrated in A375 melanoma cells. CD exhibited thiol reactivity as it was able to directly interact with N-acetylcysteine (NAC) in a cell-free assay and to induce heme oxygenase-1 (HO-1) in all cell types. And that took place in a reactive oxygen species (ROS)-independent manner. DOX decreased the mitochondrial membrane potential (Δψm) in all cell types, whereas CD selectively decreased mitochondrial respiration, affecting basal respiration, maximal respiration, spare respiratory capacity and ATP production in A375 melanoma cells, but not in healthy cardiac myoblasts. The DOX-induced cytotoxicity seen in melanoma cells was ROS-independent, whereas the cytotoxic effect of CD was associated with CD-induced ROS-formation and/or its thiol reactivity. This study highlights the beneficial properties of the addition of CD to DOX treatment, which might protect patients from DOX-induced cardiotoxicity. Future experiments with other tumor cell lines or a mouse model should substantiate this hypothesis.
Collapse
Affiliation(s)
- Lara Ebbert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany (C.-K.W.)
| | | | | | | | | | - Peter Brenneisen
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany (C.-K.W.)
| |
Collapse
|
3
|
Hsieh DJY, Tsai BCK, Barik P, Shibu MA, Kuo CH, Kuo WW, Lin PY, Shih CY, Lin SZ, Ho TJ, Huang CY. Human adipose-derived stem cells preconditioned with a novel herbal formulation Jing Shi attenuate doxorubicin-induced cardiac damage. Aging (Albany NY) 2023; 15:9167-9181. [PMID: 37708248 PMCID: PMC10522400 DOI: 10.18632/aging.205026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/21/2023] [Indexed: 09/16/2023]
Abstract
Pathological cardiac hypertrophy is a considerable contributor to global disease burden. Chinese herbal medicine (CHM) has been used to treat cardiovascular diseases since antiquity. Enhancing stem cell-mediated recovery through CHM represents a promising approach for protection against doxorubicin (Dox)-induced cardiac hypertrophy. Herein, we investigated whether human adipose-derived stem cells (hADSCs) preconditioned with novel herbal formulation Jing Si (JS) improved protective ability of stem cells against doxorubicin-induced cardiac damage. The effect of JS on hADSC viability and migration capacity was determined via MTT and migration assays, respectively. Co-culture of hADSC or JS-preconditioned hADSCs with H9c2 cells was analyzed with immunoblot, flow cytometry, TUNEL staining, LC3B staining, F-actin staining, and MitoSOX staining. The in vivo study was performed M-mode echocardiography after the treatment of JS and JS-preconditioned hADSCs by using Sprague Dawley (SD) rats. Our results indicated that JS at doses below 100 μg/mL had less cytotoxicity in hADSC and JS-preconditioned hADSCs exhibited better migration. Our results also revealed that DOX enhanced apoptosis, cardiac hypertrophy, and mitochondrial reactive oxygen species in DOX-challenged H9c2 cells, while H9c2 cells co-cultured with JS-preconditioned hADSCs alleviated these effects. It also enhanced the expression of autophagy marker LC3B, mTOR and CHIP in DOX-challenged H9c2 cells after co-culture with JS-preconditioned hADSCs. In Dox-challenged rats, the ejection fraction and fractional shortening improved in DOX-challenged SD rats exposed to JS-preconditioned hADSCs. Taken together, our data indicate that JS-preconditioned stem cells exhibit a cardioprotective capacity both in vitro and in vivo, highlighting the value of this therapeutic approach for regenerative therapy.
Collapse
Affiliation(s)
- Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Bruce Chi-Kang Tsai
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Parthasarathi Barik
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | | | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
- Department of Kinesiology and Health Science, College of William and Mary, Williamsburg, USA
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
| | - Pi-Yu Lin
- Buddhist Compassion Relief Tzu Chi Foundation, Hualien, Taiwan
| | | | - Shinn-Zong Lin
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Tsung-Jung Ho
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Hualien, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung City, Taiwan
| |
Collapse
|
4
|
Macrì R, Bava I, Scarano F, Mollace R, Musolino V, Gliozzi M, Greco M, Foti D, Tucci L, Maiuolo J, Carresi C, Tavernese A, Palma E, Muscoli C, Mollace V. In Vitro Evaluation of Ferutinin Rich- Ferula communis L., ssp. glauca, Root Extract on Doxorubicin-Induced Cardiotoxicity: Antioxidant Properties and Cell Cycle Modulation. Int J Mol Sci 2023; 24:12735. [PMID: 37628916 PMCID: PMC10454821 DOI: 10.3390/ijms241612735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
The clinical use of anthracycline Doxorubicin as an antineoplastic drug in cancer therapy is limited by cardiotoxic effects that can lead to congestive heart failure. Recent studies have shown several promising activities of different species of the genus Ferula belonging to the Apiaceae Family. Ferula communis is the main source of Ferutinin-a bioactive compound isolated from many species of Ferula-studied both in vitro and in vivo because of their different effects, such as estrogenic, antioxidant, anti-inflammatory, and also antiproliferative and cytotoxic activity, performed in a dose-dependent and cell-dependent way. However, the potential protective role of Ferutinin in myocardium impairment, caused by chemotherapeutic drugs, still represents an unexplored field. The aim of this study was to test the effects of Ferutinin rich-Ferula communis L. root extract (FcFE) at different concentrations on H9C2 cells. Moreover, we evaluated its antioxidant properties in cardiomyocytes in order to explore new potential therapeutic activities never examined before in other experimental works. FcFE, at a concentration of 0.25 µM, in the H9C2 line, significantly reduced the ROS production induced by H2O2 (50 µM and 250 µM) and traced the cell mortality of the H9C2 co-treated with Ferutinin 0.25 µM and Doxorubicin (0.5 µM and 1 µM) to control levels. These results showed that FcFE could protect against Doxorubicin-induced cardiotoxicity. Further molecular characterization of this natural compound may open the way for testing FcFE at low concentrations in vivo and in clinical studies as an adjuvant in cancer therapy in association with anthracyclines to prevent side effects on heart cells.
Collapse
Affiliation(s)
- Roberta Macrì
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Irene Bava
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Federica Scarano
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Rocco Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy
- Division of Cardiology, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy
| | - Vincenzo Musolino
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (J.M.)
| | - Micaela Gliozzi
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Marta Greco
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (D.F.)
| | - Daniela Foti
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (M.G.); (D.F.)
| | - Luigi Tucci
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Jessica Maiuolo
- Pharmaceutical Biology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (J.M.)
| | - Cristina Carresi
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (C.C.); (E.P.)
| | - Annamaria Tavernese
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Ernesto Palma
- Veterinary Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (C.C.); (E.P.)
| | - Carolina Muscoli
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
| | - Vincenzo Mollace
- Pharmacology Laboratory, Institute of Research for Food Safety and Health IRC-FSH, Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (I.B.); (M.G.); (L.T.); (A.T.); (C.M.); (V.M.)
- Renato Dulbecco Institute, Lamezia Terme, 88046 Catanzaro, Italy
| |
Collapse
|
5
|
Pharoah BM, Zhang C, Khodade VS, Keceli G, McGinity C, Paolocci N, Toscano JP. Hydropersulfides (RSSH) attenuate doxorubicin-induced cardiotoxicity while boosting its anticancer action. Redox Biol 2023; 60:102625. [PMID: 36773545 PMCID: PMC9929489 DOI: 10.1016/j.redox.2023.102625] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/19/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Cardiotoxicity is a frequent and often lethal complication of doxorubicin (DOX)-based chemotherapy. Here, we report that hydropersulfides (RSSH) are the most effective reactive sulfur species in conferring protection against DOX-induced toxicity in H9c2 cardiac cells. Mechanistically, RSSH supplementation alleviates the DOX-evoked surge in reactive oxygen species (ROS), activating nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent pathways, thus boosting endogenous antioxidant defenses. Simultaneously, RSSH turns on peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), a master regulator of mitochondrial function, while decreasing caspase-3 activity to inhibit apoptosis. Of note, we find that RSSH potentiate anticancer DOX effects in three different cancer cell lines, with evidence that suggests this occurs via induction of reductive stress. Indeed, cancer cells already exhibit much higher basal hydrogen sulfide (H2S), sulfane sulfur, and reducing equivalents compared to cardiac cells. Thus, RSSH may represent a new promising avenue to fend off DOX-induced cardiotoxicity while boosting its anticancer effects.
Collapse
Affiliation(s)
- Blaze M Pharoah
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Chengximeng Zhang
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Vinayak S Khodade
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Gizem Keceli
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Christopher McGinity
- Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Department of Biomedical Sciences, University of Padova, Padova, Italy.
| | - John P Toscano
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, United States.
| |
Collapse
|
6
|
Hwang S, Kim SH, Yoo KH, Chung MH, Lee JW, Son KH. Exogenous 8-hydroxydeoxyguanosine attenuates doxorubicin-induced cardiotoxicity by decreasing pyroptosis in H9c2 cardiomyocytes. BMC Mol Cell Biol 2022; 23:55. [DOI: 10.1186/s12860-022-00454-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
AbstractDoxorubicin (DOX), which is widely used in cancer treatment, can induce cardiomyopathy. One of the main mechanisms whereby DOX induces cardiotoxicity involves pyroptosis through the NLR family pyrin domain containing 3 (NLRP3) inflammasome and gasdermin D (GSDMD). Increased NAPDH oxidase (NOX) and oxidative stress trigger pyroptosis. Exogenous 8-hydroxydeoxyguanosine (8-OHdG) decreases reactive oxygen species (ROS) production by inactivating NOX. Here, we examined whether 8-OHdG treatment can attenuate DOX-induced pyroptosis in H9c2 cardiomyocytes. Exposure to DOX increased the peroxidative glutathione redox status and NOX1/2/4, toll-like receptor (TLR)2/4, and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) expression, while an additional 8-OHdG treatment attenuated these effects. Furthermore, DOX induced higher expression of NLRP3 inflammasome components, including NLRP3, apoptosis-associated speck-like protein containing a c-terminal caspase recruitment domain (ASC), and pro-caspase-1. Moreover, it increased caspase-1 activity, a marker of pyroptosis, and interleukin (IL)-1β expression. All these effects were attenuated by 8-OHdG treatment. In addition, the expression of the cardiotoxicity markers, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) was increased by DOX, whereas the increase of ANP and BNP induced by DOX treatment was reversed by 8-OHdG. In conclusion, exogenous 8-OHdG attenuated DOX-induced pyroptosis by decreasing the expression of NOX1/2/3, TLR2/4, and NF-κB. Thus, 8-OHdG may attenuate DOX-induced cardiotoxicity through the inhibition of pyroptosis.
Collapse
|
7
|
Martins B, Novo JP, Fonseca É, Raposo R, Sardão VA, Pereira F, Oriá RB, Fontes-Ribeiro C, Malva J. Necrotic-like BV-2 microglial cell death due to methylmercury exposure. Front Pharmacol 2022; 13:1003663. [PMID: 36408241 PMCID: PMC9667718 DOI: 10.3389/fphar.2022.1003663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/18/2022] [Indexed: 12/01/2022] Open
Abstract
Methylmercury (MeHg) is a dangerous environmental contaminant with strong bioaccumulation in the food chain and neurotoxic properties. In the nervous system, MeHg may cause neurodevelopment impairment and potentially interfere with immune response, compromising proper control of neuroinflammation and aggravating neurodegeneration. Human populations are exposed to environmental contamination with MeHg, especially in areas with strong mining or industrial activity, raising public health concerns. Taking this into consideration, this work aims to clarify pathways leading to acute toxic effects caused by MeHg exposure in microglial cells. BV-2 mouse microglial cells were incubated with MeHg at different concentrations (0.01, 0.1, 1 and 10 µM) for 1 h prior to continuous Lipopolysaccharide (LPS, 0.5 μg/ml) exposure for 6 or 24 h. After cell exposure, reactive oxygen species (ROS), IL-6 and TNF-α cytokines production, inducible nitric oxide synthase (iNOS) expression, nitric oxide (NO) release, metabolic activity, propidium iodide (PI) uptake, caspase-3 and -9 activities and phagocytic activity were assessed. MeHg 10 µM decreased ROS formation, the production and secretion of pro-inflammatory cytokines IL-6, TNF-α, iNOS immunoreactivity, the release of NO in BV-2 cells. Furthermore, MeHg 10 µM decreased the metabolic activity of BV-2 and increased the number of PI-positive cells (necrotic-like cell death) when compared to the respective control group. Besides, MeHg did not interfere with caspase activity or the phagocytic profile of cells. The short-term effects of a high concentration of MeHg on BV-2 microglial cells lead to impaired production of several pro-inflammatory mediators, as well as a higher microglial cell death via necrosis, compromising their neuroinflammatory response. Clarifying the mechanisms underlying MeHg-induced neurotoxicity and neurodegeneration in brain cells is relevant to better understand acute and long-term chronic neuroinflammatory responses following MeHg exposure.
Collapse
Affiliation(s)
- B. Martins
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - J. P. Novo
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - É. Fonseca
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - R. Raposo
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal,Experimental Biology Core, Health Sciences Center, University of Fortaleza, Fortaleza, Brazil
| | - V. A. Sardão
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal,Center for Neuroscience and Cell Biology (CNC), UC Biotech, University of Coimbra, Cantanhede, Portugal
| | - F. Pereira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - R. B. Oriá
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology and Institute of Biomedicine, School of Medicine, Federal University of Ceará, Fortaleza, Brazil
| | - C. Fontes-Ribeiro
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - J. Malva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal,Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal,Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal,*Correspondence: J. Malva,
| |
Collapse
|
8
|
Liao W, Rao Z, Wu L, Chen Y, Li C. Cariporide Attenuates Doxorubicin-Induced Cardiotoxicity in Rats by Inhibiting Oxidative Stress, Inflammation and Apoptosis Partly Through Regulation of Akt/GSK-3β and Sirt1 Signaling Pathway. Front Pharmacol 2022; 13:850053. [PMID: 35747748 PMCID: PMC9209753 DOI: 10.3389/fphar.2022.850053] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Doxorubicin (DOX) is a potent chemotherapeutic agent with limited usage due to its cumulative cardiotoxicity. The Na+/H+ exchanger isoform 1 (NHE1) is a known regulator of oxidative stress, inflammation, and apoptosis. The present study was designed to investigate the possible protective effect of cariporide (CAR), a selective inhibitor of NHE1, against DOX-induced cardiotoxicity in rats. Methods: Male Sprague-Dawley rats were intraperitoneally injected with DOX to induce cardiac toxicity and CAR was given orally for treatment. The injured H9c2 cell model was established by incubation with DOX in vitro. Echocardiography, as well as morphological and ultra-structural examination were performed to evaluate cardiac function and histopathological changes. The biochemical parameters were determined according to the manufacturer’s guideline of kits. ROS were assessed by using an immunofluorescence assay. The serum levels and mRNA expressions of inflammatory cytokines were measured by using ELISA or qRT-PCR. Cardiac cell apoptosis and H9c2 cell viability were tested by TUNEL or MTT method respectively. The protein expressions of Cleaved-Caspase-3, Bcl-2, Bax, Akt, GSK-3β, and Sirt1 were detected by western blot. Results: Treatment with CAR protected against DOX-induced body weight changes, impairment of heart function, leakage of cardiac enzymes, and heart histopathological damage. In addition, CAR significantly attenuated oxidative stress and inhibited the levels and mRNA expressions of inflammatory cytokines (TNF-α, IL-6, IL-18, and IL-1β), which were increased by DOX treatment. Moreover, CAR significantly suppressed myocardial apoptosis and Cleaved-Caspase-3 protein expression induced by DOX, which was in agreement with the increased Bcl-2/Bax ratio. Also, DOX suppressed phosphorylation of Akt and GSK-3β, which was significantly reversed by administration of CAR. Furthermore, CAR treatment prevented DOX-induced down-regulation of Sirt1 at the protein level in vitro and in vivo. Finally, Sirt1 inhibitor reversed the protective effects of CAR, as evidenced by reduced cell viability and Sirt1 protein expression in vitro. Conclusion: Taken together, we provide evidence for the first time in the current study that CAR exerts potent protective effects against DOX-induced cardiotoxicity in rats. This cardio-protective effect is attributed to suppressing oxidative stress, inflammation, and apoptosis, at least in part, through regulation of Akt/GSK-3β and Sirt1 signaling pathway, which has not been reported to date.
Collapse
Affiliation(s)
- Wenli Liao
- National Demonstration Center for Experimental General Medicine Education, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhiwei Rao
- Central Hospital of Xianning, The First Affiliate Hospital of Hubei University of Science and Technology, Xianning, China
| | - Lingling Wu
- National Demonstration Center for Experimental General Medicine Education, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yong Chen
- Central Hospital of Xianning, The First Affiliate Hospital of Hubei University of Science and Technology, Xianning, China
- *Correspondence: Cairong Li, ; Yong Chen,
| | - Cairong Li
- National Demonstration Center for Experimental General Medicine Education, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
- *Correspondence: Cairong Li, ; Yong Chen,
| |
Collapse
|
9
|
Schwaerzer GK, Casteel DE, Cividini F, Kalyanaraman H, Zhuang S, Gu Y, Peterson KL, Dillmann W, Boss GR, Pilz RB, Pilz RB. Constitutive protein kinase G activation exacerbates stress-induced cardiomyopathy. Br J Pharmacol 2022; 179:2413-2429. [PMID: 34000062 PMCID: PMC9926932 DOI: 10.1111/bph.15530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 04/29/2021] [Accepted: 05/04/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Heart failure is associated with high morbidity and mortality, and new therapeutic targets are needed. Preclinical data suggest that pharmacological activation of protein kinase G (PKG) can reduce maladaptive ventricular remodelling and cardiac dysfunction in the stressed heart. However, clinical trial results have been mixed and the effects of long-term PKG activation in the heart are unknown. EXPERIMENTAL APPROACH We characterized the cardiac phenotype of mice carrying a heterozygous knock-in mutation of PKG1 (Prkg1R177Q/+ ), which causes constitutive, cGMP-independent activation of the kinase. We examined isolated cardiac myocytes and intact mice, the latter after stress induced by surgical transaortic constriction or angiotensin II (Ang II) infusion. KEY RESULTS Cardiac myocytes from Prkg1R177Q/+ mice showed altered phosphorylation of sarcomeric proteins and reduced contractility in response to electrical stimulation, compared to cells from wild type mice. Under basal conditions, young PKG1R177Q/+ mice exhibited no obvious cardiac abnormalities, but aging animals developed mild increases in cardiac fibrosis. In response to angiotensin II infusion or fixed pressure overload induced by transaortic constriction, young PKGR177Q/+ mice exhibited excessive hypertrophic remodelling with increased fibrosis and myocyte apoptosis, leading to increased left ventricular dilation and dysfunction compared to wild type litter mates. CONCLUSION AND IMPLICATIONS Long-term PKG1 activation in mice may be harmful to the heart, especially in the presence of pressure overload and neurohumoral stress. LINKED ARTICLES This article is part of a themed issue on cGMP Signalling in Cell Growth and Survival. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v179.11/issuetoc.
Collapse
Affiliation(s)
- Gerburg K. Schwaerzer
- Department of Medicine, University of California San Diego, La Jolla, California 92093,These three authors contributed equally to the work
| | - Darren E. Casteel
- Department of Medicine, University of California San Diego, La Jolla, California 92093,These three authors contributed equally to the work
| | - Federico Cividini
- Department of Medicine, University of California San Diego, La Jolla, California 92093,These three authors contributed equally to the work
| | - Hema Kalyanaraman
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Shunhui Zhuang
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Yusu Gu
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Kirk L. Peterson
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Wolfgang Dillmann
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Gerry R. Boss
- Department of Medicine, University of California San Diego, La Jolla, California 92093
| | - Renate B. Pilz
- Department of Medicine, University of California San Diego, La Jolla, California 92093,Correspondence should be addressed to R.B.P. ()
| | - Renate B Pilz
- Department of Medicine, University of California San Diego, La Jolla, California, 92093, USA
| |
Collapse
|
10
|
Chang WT, Shih JY, Lin YW, Chen ZC, Kan WC, Lin TH, Hong CS. Dapagliflozin protects against doxorubicin-induced cardiotoxicity by restoring STAT3. Arch Toxicol 2022; 96:2021-2032. [PMID: 35438302 DOI: 10.1007/s00204-022-03298-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/31/2022] [Indexed: 01/02/2023]
Abstract
Doxorubicin (Dox), an effective therapy in different types of cancer, is known to exhibit cardiotoxic effects. Despite previous studies indicating the benefits of dapagliflozin (DAPA) in patients experiencing heart failure, it remains uncertain whether DAPA exerts a protective effect on Dox-induced cardiac dysfunction. Signal transducer and activator of transcription 3 (STAT3) participates in various mechanisms of cardioprotection. Herein, we aimed to investigate the effects of DAPA on Dox-induced cardiotoxicity and the role of STAT3. Sprague-Dawley rats were pretreated with oral DAPA for 6 weeks followed by Dox for 4 weeks. Sequential echocardiography was applied to assess cardiac function. For in vitro analysis, cardiomyocytes were treated with 10 μM DAPA and subsequently exposed to 1 μM Dox. The expression of reactive oxygen species- and apoptosis-related proteins was measured. Using STAT3 siRNA, we further examined the effects of STAT3 effect on DAPA-associated protection against Dox-induced apoptosis. In rats treated with Dox, DAPA significantly reduced cardiac fibrosis and improved cardiac function and hemodynamics. Additionally, DAPA effectively inhibited Dox-induced apoptosis and reactive oxygen species (ROS) in cardiomyocytes. Mechanistically, we showed that DAPA decreased cardiac expression of Bax and cleaved caspase 3 but increased Bcl-2 expression. DAPA also significantly rescued Dox-suppressed STAT3 expression. Conversely, knocking down STAT3 in cardiomyocytes reversed the DAPA-related protective effects on Dox-induced cell apoptosis and ROS. Collectively, our findings indicate that DAPA could be useful for preventing Dox-induced cardiotoxicity by restoring STAT3.
Collapse
Affiliation(s)
- Wei-Ting Chang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Jhih-Yuan Shih
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Yu-Wen Lin
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Zhih-Cherng Chen
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Wei-Chih Kan
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, 901, Zhonghua Road, Yongkang District, Tainan, Taiwan ROC.
- Department of Biological Science and Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan.
| | - Tsung-Hsien Lin
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Chon-Seng Hong
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| |
Collapse
|
11
|
Kitakata H, Endo J, Ikura H, Moriyama H, Shirakawa K, Katsumata Y, Sano M. Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis. Int J Mol Sci 2022; 23:1414. [PMID: 35163335 PMCID: PMC8835899 DOI: 10.3390/ijms23031414] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 01/04/2023] Open
Abstract
Doxorubicin (DOX) is the most widely used anthracycline anticancer agent; however, its cardiotoxicity limits its clinical efficacy. Numerous studies have elucidated the mechanisms underlying DOX-induced cardiotoxicity, wherein apoptosis has been reported as the most common final step leading to cardiomyocyte death. However, in the past two years, the involvement of ferroptosis, a novel programmed cell death, has been proposed. The purpose of this review is to summarize the historical background that led to each form of cell death, focusing on DOX-induced cardiotoxicity and the molecular mechanisms that trigger each form of cell death. Furthermore, based on this understanding, possible therapeutic strategies to prevent DOX cardiotoxicity are outlined. DNA damage, oxidative stress, intracellular signaling, transcription factors, epigenetic regulators, autophagy, and metabolic inflammation are important factors in the molecular mechanisms of DOX-induced cardiomyocyte apoptosis. Conversely, the accumulation of lipid peroxides, iron ion accumulation, and decreased expression of glutathione and glutathione peroxidase 4 are important in ferroptosis. In both cascades, the mitochondria are an important site of DOX cardiotoxicity. The last part of this review focuses on the significance of the disruption of mitochondrial homeostasis in DOX cardiotoxicity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan; (H.K.); (J.E.); (H.I.); (H.M.); (K.S.); (Y.K.)
| |
Collapse
|
12
|
Yao T, Fujimura T, Murayama K, Okumura K, Seko Y. Oxidative stress-responsive apoptosis inducing protein (ORAIP) plays a critical role in doxorubicin-induced apoptosis in rat cardiac myocytes. Int J Cardiol 2021; 348:119-124. [PMID: 34864083 DOI: 10.1016/j.ijcard.2021.11.085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Oxidative stress is implicated in the pathogenesis of doxorubicin-induced apoptosis in cardiac myocytes. However, the precise mechanism remains uncertain. We identified an apoptosis-inducing humoral factor, in a conditioned medium from cardiac myocytes subjected to hypoxia/reoxygenation, to be 69th tyrosine-sulfated eukaryotic translation initiation factor 5A (eIF5A). We named this novel secreted form of eIF5A, Oxidative stress-Responsive Apoptosis Inducing Protein (ORAIP). We confirmed that ischemia/reperfusion, ultraviolet-irradiation, and ionizing radiation significantly increased plasma levels of ORAIP in vivo, supporting that secretion of ORAIP is specific to the oxidative stress. To investigate the role of ORAIP in doxorubicin-induced apoptosis of cardiac myocytes. METHODS We analyzed plasma levels of ORAIP in rats treated with doxorubicin (10 mg/Kg) in vivo, and the effects of neutralizing anti-ORAIP monoclonal antibody (mAb) on doxorubicin-induced apoptosis of cardiac myocytes in vitro. RESULTS The (mean ± SE) plasma ORAIP levels before doxorubicin administration were (13.7 ± 2.7) ng/mL, they markedly increased with peak levels ([178.6 ± 6.5] ng/mL, p < 0.00001, vs. before administration) at 20 to 60 min after doxorubicin administration, then gradually decreased to (118.0 ± 4.8) ng/mL at 120 min. Treatment with a neutralizing anti-ORAIP mAb significantly (nearly 50%) suppressed doxorubicin-induced apoptosis of cardiac myocytes. CONCLUSIONS These data indicate that doxorubicin induces oxidative stress resulting in the strong expression of ORAIP in cardiac myocytes and marked secretion of ORAIP into peripheral circulation. This strongly suggests that ORAIP can be a novel sensitive biomarker as well as a possible therapeutic target for doxorubicin-induced cell injury in anti-cancer therapy.
Collapse
Affiliation(s)
- Takako Yao
- Division of Cardiovascular Medicine, Institute for Adult Diseases, Asahi Life Foundation, Tokyo 103-0002, Japan
| | - Tsutomu Fujimura
- Laboratory of Bioanalytical Chemistry, Tohoku Medical and Pharmaceutical University, Sendai 981-0905, Japan
| | - Kimie Murayama
- Division of Proteomics and Biomolecular Science, BioMedical Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Ko Okumura
- Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Yoshinori Seko
- Department of Biofunctional Microbiota, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan.
| |
Collapse
|
13
|
Calpain-Mediated Mitochondrial Damage: An Emerging Mechanism Contributing to Cardiac Disease. Cells 2021; 10:cells10082024. [PMID: 34440793 PMCID: PMC8392834 DOI: 10.3390/cells10082024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/19/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Calpains belong to the family of calcium-dependent cysteine proteases expressed ubiquitously in mammals and many other organisms. Activation of calpain is observed in diseased hearts and is implicated in cardiac cell death, hypertrophy, fibrosis, and inflammation. However, the underlying mechanisms remain incompletely understood. Recent studies have revealed that calpains target and impair mitochondria in cardiac disease. The objective of this review is to discuss the role of calpains in mediating mitochondrial damage and the underlying mechanisms, and to evaluate whether targeted inhibition of mitochondrial calpain is a potential strategy in treating cardiac disease. We expect to describe the wealth of new evidence surrounding calpain-mediated mitochondrial damage to facilitate future mechanistic studies and therapy development for cardiac disease.
Collapse
|
14
|
Jadhao M, Tsai EM, Yang HC, Chen YF, Liang SS, Wang TN, Teng YN, Huang HW, Wang LF, Chiu CC. The Long-Term DEHP Exposure Confers Multidrug Resistance of Triple-Negative Breast Cancer Cells through ABC Transporters and Intracellular ROS. Antioxidants (Basel) 2021; 10:949. [PMID: 34208283 PMCID: PMC8230873 DOI: 10.3390/antiox10060949] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/18/2022] Open
Abstract
The characteristics of phthalates had been thought to be similar to endocrine disruptors, which increases cancer risk. The role of phthalates in acquired drug resistance remains unclear. In this study, we investigated the effect of di-(2-ethylhexyl) phthalate (DEHP) on acquired drug resistance in breast cancer. MCF7 and MDA-MB-231 breast cancer cells were exposed to long-term physiological concentration of DEHP for more than three months. Long-exposure DEHP permanently attenuated the anti-proliferative effect of doxorubicin with estrogen receptor-independent activity even after withdrawal of DEHP. Long term DEHP exposure significantly reduced ROS (O2-) level in MDA-MB-231 cells while increased in MCF7 cells. ATP-binding cassette (ABC) transporters possess a widely recognized mechanism of drug resistance and are considered a target for drug therapy. Upregulation of ABC family proteins, ABCB-1 and ABCC-1 observed in DEHP-exposed clones compared to doxorubicin-resistant (DoxR) and parental MDA-MB-231 cells. A viability assay showed enhanced multidrug resistance in DEHP-exposed clones against Dox, topotecan, and irinotecan. Inhibition of ABC transporters with tariquidar, enhanced drug cytotoxicity through increased drug accumulation reversing acquired multidrug resistance in MDA-MB-231 breast cancer cells. Tariquidar enhanced Dox cytotoxicity by increasing intracellular ROS production leading to caspase-3 mediated apoptosis. Activation of PI3K/Akt signaling enhanced proliferation and growth of DEHP-exposed MDA-MB-231 cells. Overall, long-term DEHP exposure resulted in acquired multidrug resistance by upregulating ABCB-1 and ABCC1; apart from proliferation PI3K/Akt may be responsible for acquired drug resistance through ABC transporter upregulation. Targeting ABCB1 and ABCC1 with tariquidar may be a promising strategy for reversing the acquired multidrug resistance of triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Mahendra Jadhao
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; or
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ho-Chun Yang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.Y.); (S.-S.L.)
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
| | - Yih-Fung Chen
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shih-Shin Liang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.Y.); (S.-S.L.)
| | - Tsu-Nai Wang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan;
| | - Hurng-Wern Huang
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan;
| | - Li-Fang Wang
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan; or
| | - Chien-Chih Chiu
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.Y.); (S.-S.L.)
- Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
15
|
Palkina KA, Ipatova DA, Shakhova ES, Balakireva AV, Markina NM. Therapeutic Potential of Hispidin-Fungal and Plant Polyketide. J Fungi (Basel) 2021; 7:jof7050323. [PMID: 33922000 PMCID: PMC8143579 DOI: 10.3390/jof7050323] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/19/2022] Open
Abstract
There is a large number of bioactive polyketides well-known for their anticancer, antibiotic, cholesterol-lowering, and other therapeutic functions, and hispidin is among them. It is a highly abundant secondary plant and fungal metabolite, which is investigated in research devoted to cancer, metabolic syndrome, cardiovascular, neurodegenerative, and viral diseases. This review summarizes over 20 years of hispidin studies of its antioxidant, anti-inflammatory, anti-apoptotic, antiviral, and anti-cancer cell activity.
Collapse
Affiliation(s)
- Kseniia A. Palkina
- Department of Biomolecular Chemistry, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (K.A.P.); (D.A.I.); (E.S.S.); (A.V.B.)
- Planta LLC, 121205 Moscow, Russia
| | - Daria A. Ipatova
- Department of Biomolecular Chemistry, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (K.A.P.); (D.A.I.); (E.S.S.); (A.V.B.)
- School of Pharmacy, Faculty of Biomedicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Ekaterina S. Shakhova
- Department of Biomolecular Chemistry, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (K.A.P.); (D.A.I.); (E.S.S.); (A.V.B.)
- Planta LLC, 121205 Moscow, Russia
| | - Anastasia V. Balakireva
- Department of Biomolecular Chemistry, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (K.A.P.); (D.A.I.); (E.S.S.); (A.V.B.)
- Planta LLC, 121205 Moscow, Russia
| | - Nadezhda M. Markina
- Department of Biomolecular Chemistry, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, 117997 Moscow, Russia; (K.A.P.); (D.A.I.); (E.S.S.); (A.V.B.)
- Planta LLC, 121205 Moscow, Russia
- Correspondence: ; Tel.: +7-9161342855
| |
Collapse
|
16
|
Potential Therapeutic Effects of New Ruthenium (III) Complex with Quercetin: Characterization, Structure, Gene Regulation, and Antitumor and Anti-Inflammatory Studies (RuIII/Q Novel Complex Is a Potent Immunoprotective Agent). CRYSTALS 2021. [DOI: 10.3390/cryst11040367] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The aim of this study was to evaluate the antioxidant and anti-inflammatory effects of the new [Ru(Q)(Cl)2(H2O)2] complex (RuIII/Q). A new vital complex containing quercetin flavonoid compound (Q) with ruthenium (III) ions was synthesized. The molar conductivity of the RuIII/Q complex was measured in dimethylsulfoxide (DMSO) with value 12 (Ω−1 mol−1 cm−1, indicating their non-electrolytic nature. Infrared (FTIR) spectroscopic investigation of the RuIII/Q complex indicated that Q is coordinated as a bidentate with Ru metal ions through the oxygen of carbonyl C(4)=O group and oxygen of phenolic C(3)−O group based on the wavenumber shifts at 1654 and 1335 cm−1 respectively. The electronic (UV−Vis) spectra and the magnetic susceptibility value (1.85 B.M.) revealed that the Ru(III) complex has an octahedral geometry. The average diameter of the RuIII/Q nanoparticles was approximately 7–15 nm according to the transmission electron microscopy. The thermogravimetric study (TG/DTG) indicates that the RuIII/Q compound is quite stable until 300 °C. To assess biological activity, 60 male rats were allocated to six groups, namely control, DG (D-galactose), Q, RuIII/Q, DG plus Q, and DG plus RuIII/Q. Antioxidant enzymes (SOD, CAT, GPx, and GRx), markers of lipid peroxidation (such as MDA), expression of genes (namely Nrf2, Cu-ZnSOD, CAT, GPx, cyto c, P53, Bax, BCl2, caspase-3, and caspase-9 in testicular tissue), glutamate, 4-hydroxynonenal (HNE), GSH, HCY, amyloid beta, and GABA levels were evaluated in brain tissues. Cytokines, such as IL-6 and TNF-α, histological and ultrastructural studies were estimated in both the brain and testicular tissues, while the comet assay was performed in the brain tissue. RuIII/Q administration either alone or combined with DG reduced oxidative injury to normal levels and decreased apoptotic activities. Thus, RuIII/Q inhibited injury in both the testis and brain and reduced oxidative stress in male rats. The (RuIII/Q) complex has a potent ameliorative effect against aging neurotoxicity, reproductive toxicity, and antihepatic cancer activity induced by D-galactose (DG).
Collapse
|
17
|
Afrostyrax lepidophyllus Mildbr. and Monodora myristica (Gaertn.) Dunal Extracts Decrease Doxorubicin Cytotoxicity on H9c2 Cardiomyoblasts. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8858165. [PMID: 33688366 PMCID: PMC7920721 DOI: 10.1155/2021/8858165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/25/2021] [Accepted: 02/05/2021] [Indexed: 11/17/2022]
Abstract
Materials and Methods Bark extracts of these plants (1 and 25 µg/mL) were added 3 hours before coincubating H9c2 cardiomyoblasts with Dox (0.5 and 1 µM) for 24 hours more. We measured cell mass and metabolic viability, mitochondrial transmembrane potential, superoxide anion content, and activity-like of caspase-3 and caspase-9 following treatment with the extracts and/or Dox. Also, selenium and vitamin C contents were measured in the plant extracts. Results The results confirmed that Dox treatment decreased cell mass, mitochondrial membrane potential and metabolic viability, increased mitochondrial superoxide anion, and stimulated caspase-3 and caspase-9-like activities. Pretreatment of the cells with the plant extracts significantly inhibited Dox cytotoxicity, with more significant results at the higher concentration. Measurements of selenium and vitamin C in the extracts revealed higher concentration of both when compared with other Cameroonian spices. Conclusion Both extracts of A. lepidophyllus and M. myristica were effective against Dox-induced cytotoxicity, most likely due to their content in antioxidants.
Collapse
|
18
|
Dong K, Zhao ZZ, Kang J, Lin LR, Chen WT, Liu JX, Wu XL, Lu TL. Cinnamaldehyde and Doxorubicin Co-Loaded Graphene Oxide Wrapped Mesoporous Silica Nanoparticles for Enhanced MCF-7 Cell Apoptosis. Int J Nanomedicine 2020; 15:10285-10304. [PMID: 33376322 PMCID: PMC7756203 DOI: 10.2147/ijn.s283981] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
Background Combined chemotherapy is often affected by the different physicochemical properties of chemotherapeutic drugs, which should be improved by the reasonable design of co-loaded preparations. Purpose A kind of simple but practical graphene oxide (GO) wrapped mesoporous silica nanoparticles (MSN) modified with hyaluronic acid (MSN@GO-HA) were developed for the co-delivery of cinnamaldehyde (CA) and doxorubicin (DOX), in order to enhance their combined treatment on tumor cells and reduce their application defects. Methods The MSNCA@GODOX-HA was constructed by MSNCA (loading CA via physical diffusion) and GODOX-HA (modified with HA and loading DOX via π–π stacking) through the electrostatic adsorption, followed by the physicochemical characterization, serum stability and in vitro release study. Cytotoxicity on different cells was detected, followed by the tumor cell uptake tests. The intracellular reactive oxygen species (ROS) changes, mitochondrial functions and activities of caspase-3/-9 in MCF-7 cells were also evaluated, respectively. Results The MSNCA@GODOX-HA nanoparticles kept stable in FBS solution and achieved pH-responsive release behavior, which was beneficial to increase the accumulation of CA and DOX in tumor cells to enhance the treatment. MSNCA@GODOX-HA exerted higher cytotoxicity to MCF-7 human breast cancer cells than H9c2 cardiac myocyte cells, which were not only attributed to the active targeting to tumor cells by HA, but also related with the activation of intrinsic apoptotic pathway in MCF-7 cells induced by CA, which was mediated by the specific ROS signal amplification and the interference with mitochondrial function. Moreover, the efficacy of DOX was also enhanced by the above process. Conclusion The establishment of the MSNCA@GODOX-HA nanoparticles played a role in promoting strengths and restricting shortcomings of CA and DOX, thereby exerting their function and achieving efficient treatment against cancer.
Collapse
Affiliation(s)
- Kai Dong
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, People's Republic of China
| | - Zhuang-Zhuang Zhao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, People's Republic of China
| | - Jian Kang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, People's Republic of China
| | - Lei-Ruo Lin
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, People's Republic of China
| | - Wen-Ting Chen
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, People's Republic of China
| | - Jin-Xi Liu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, People's Republic of China
| | - Xiang-Long Wu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, People's Republic of China
| | - Ting-Li Lu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
19
|
Liu C, Cai Z, Hu T, Yao Q, Zhang L. Cathepsin B aggravated doxorubicin‑induced myocardial injury via NF‑κB signalling. Mol Med Rep 2020; 22:4848-4856. [PMID: 33173960 PMCID: PMC7646931 DOI: 10.3892/mmr.2020.11583] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 08/25/2020] [Indexed: 11/09/2022] Open
Abstract
Myocyte apoptosis and oxidative stress key critical roles in the process of doxorubicin (DOX)-induced cardiotoxicity. However, how apoptosis and oxidative stress arise in DOX-induced heart injury remains largely unknown. Cathepsin B (CTSB) is a typical lysosomal cysteine protease that is associated with apoptosis, inflammatory responses, oxidative stress and autophagy. The present study aimed to investigate the role of CTSB in DOX-induced heart injury and its potential mechanism. H9C2 cells were infected with adenovirus or transfected with small interfering RNA to overexpress or knock down CTSB, respectively, and then stimulated with DOX. DOX induced increased CTSB expression levels in H9C2 cells. DOX-induced cardiomyocyte apoptosis and oxidative stress were attenuated by CTSB knockdown but aggravated by CTSB overexpression in vitro. Mechanistically, the present study showed that CTSB activated the NF-κB pathway in response to DOX. In summary, CTSB aggravated DOX-induced H9C2 cell apoptosis and oxidative stress via NF-κB signalling. CTSB constitutes a potential therapeutic target for the treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Chen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zhulan Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tongtong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lijun Zhang
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
20
|
Dallons M, Alpan E, Schepkens C, Tagliatti V, Colet JM. GPR91 Receptor Mediates Protection against Doxorubicin-Induced Cardiotoxicity without Altering Its Anticancer Efficacy. An In Vitro Study on H9C2 Cardiomyoblasts and Breast Cancer-Derived MCF-7 Cells. Cells 2020; 9:E2177. [PMID: 32992522 PMCID: PMC7599858 DOI: 10.3390/cells9102177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 11/16/2022] Open
Abstract
Doxorubicin (DOX) is an anticancer drug widely used in oncology, especially for breast cancer. The main limitation of DOX treatment is its cardiotoxicity due to the cumulative dose. Clinically, DOX-induced cardiomyopathy develops as a progressive heart failure caused by a progressive cardiomyocyte's death. For long, the oxidative stress induced by DOX was considered as the main toxic mechanism responsible for heart damage, but it is now controverted, and other processes are investigated to develop cardioprotective strategies. Previously, we studied DOX-induced cardiotoxicity and dexrazoxane (DEX), the only cardioprotective compound authorized by the FDA, by 1H-NMR metabonomics in H9C2 cells. We observed an increased succinate secretion in the extracellular fluid of DEX-exposed cardiomyocytes, a finding that led us to the hypothesis of a possible protective role of this agonist of the GPR91 receptor. The objective of the present work was to study the effect of succinate (SUC) and cis-epoxysuccinate (cis-ES), two agonists of the GPR91 receptor, on DOX-induced cardiotoxicity to H9C2 cells. To this purpose, several toxicity parameters, including cell viability, oxidative stress and apoptosis, as well as the GPR91 expression, were measured to assess the effects of DEX, SUC and cis-ES either alone or in combination with DOX in H9C2 cells. A 1H-NMR-based metabonomic study was carried out on cellular fluids collected after 24 h to highlight the metabolic changes induced by those protective compounds. Moreover, the effects of each agonist given either alone or in combination with DOX were evaluated on MCF-7 breast cancer cells. GPR91 expression was confirmed in H9C2 cells, while no expression was found in MCF-7 cells. Under such experimental conditions, both SUC and cis-ES decreased partially the cellular mortality, the oxidative stress and the apoptosis induced by DOX. The SUC protective effect was similar to the DEX effect, but the protective effect of cis-ES was higher on oxidative stress and apoptosis. In addition, the metabonomics findings pointed out several metabolic pathways involved in the cardioprotective effects of both GPR91 agonists: the stimulation of aerobic metabolism with glucose as the main fuel, redox balance and phospholipids synthesis. Finally, none of the GPR91 agonists jeopardized the pharmacological effects of DOX on MCF-7 breast cancer cells.
Collapse
Affiliation(s)
| | | | | | | | - Jean-Marie Colet
- Department of Human Biology & Toxicology, Faculty of Medicine and Pharmacy, University of Mons, Place du Parc 20, 7000 Mons, Belgium; (M.D.); (E.A.); (C.S.); (V.T.)
| |
Collapse
|
21
|
Anthracycline-Related Heart Failure: Certain Knowledge and Open Questions : Where Do we Stand with Chemotherapyinduced Cardiotoxicity? Curr Heart Fail Rep 2020; 17:357-364. [PMID: 32964378 PMCID: PMC7683464 DOI: 10.1007/s11897-020-00489-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2020] [Indexed: 12/24/2022]
Abstract
In the last decade, cardio-oncology has become a discipline on its own, with tremendous research going on to unravel the mechanisms underpinning different manifestations of cardiotoxicity caused by anticancer drugs. Although this domain is much broader than the effect of chemotherapy alone, a lot of questions about anthracycline-induced cardiotoxicity remain unknown. In this invited review, we provide insights in molecular mechanisms behind anthracycline-induced cardiotoxicity and put it in a clinical framework emphasizing the need for patients to understand, detect, and treat this detrimental condition.
Collapse
|
22
|
Chen Q, Samidurai A, Thompson J, Hu Y, Das A, Willard B, Lesnefsky EJ. Endoplasmic reticulum stress-mediated mitochondrial dysfunction in aged hearts. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165899. [PMID: 32698045 DOI: 10.1016/j.bbadis.2020.165899] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/04/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022]
Abstract
Aging impairs the mitochondrial electron transport chain (ETC), especially in interfibrillar mitochondria (IFM). Mitochondria are in close contact with the endoplasmic reticulum (ER). Induction of ER stress leads to ETC injury in adult heart mitochondria. We asked if ER stress contributes to the mitochondrial dysfunction during aging. Subsarcolemmal mitochondria (SSM) and IFM were isolated from 3, 18, and 24 mo. C57Bl/6 mouse hearts. ER stress progressively increased with age, especially in 24 mo. mice that manifest mitochondrial dysfunction. OXPHOS was decreased in 24 mo. IFM oxidizing complex I and complex IV substrates. Proteomic analysis showed that the content of multiple complex I subunits was decreased in IFM from 24 mo. hearts, but remained unchanged in in 18 mo. IFM without a decrease in OXPHOS. Feeding 24 mo. old mice with 4-phenylbutyrate (4-PBA) for two weeks attenuated the ER stress and improved mitochondrial function. These results indicate that ER stress contributes to the mitochondrial dysfunction in aged hearts. Attenuation of ER stress is a potential approach to improve mitochondrial function in aged hearts.
Collapse
Affiliation(s)
- Qun Chen
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Arun Samidurai
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Jeremy Thompson
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Ying Hu
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Anindita Das
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America
| | - Belinda Willard
- Proteomics Core, Cleveland Clinic, Cleveland, OH 44106, United States of America
| | - Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, VA 23298, United States of America; Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, 23298, United States of America; Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, VA 23298, United States of America; McGuire Department of Veterans Affairs Medical Center, Richmond, VA 23249, United States of America.
| |
Collapse
|
23
|
Abstract
Anthracycline-based chemotherapy can result in the development of a cumulative and progressively developing cardiomyopathy. Doxorubicin is one of the most highly prescribed anthracyclines in the United States due to its broad spectrum of therapeutic efficacy. Interference with different mitochondrial processes is chief among the molecular and cellular determinants of doxorubicin cardiotoxicity, contributing to the development of cardiomyopathy. The present review provides the basis for the involvement of mitochondrial toxicity in the different functional hallmarks of anthracycline toxicity. Our objective is to understand the molecular determinants of a progressive deterioration of functional integrity of mitochondria that establishes a historic record of past drug treatments (mitochondrial memory) and renders the cancer patient susceptible to subsequent regimens of drug therapy. We focus on the involvement of doxorubicin-induced mitochondrial oxidative stress, disruption of mitochondrial oxidative phosphorylation, and permeability transition, contributing to altered metabolic and redox circuits in cardiac cells, ultimately culminating in disturbances of autophagy/mitophagy fluxes and increased apoptosis. We also suggest some possible pharmacological and nonpharmacological interventions that can reduce mitochondrial damage. Understanding the key role of mitochondria in doxorubicin-induced cardiomyopathy is essential to reduce the barriers that so dramatically limit the clinical success of this essential anticancer chemotherapy.
Collapse
Affiliation(s)
- Kendall B Wallace
- From the Department of Biomedical Sciences, University of Minnesota Medical School, Duluth (K.B.W.)
| | - Vilma A Sardão
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal (V.A.S., P.J.O.)
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal (V.A.S., P.J.O.)
| |
Collapse
|
24
|
Dallons M, Schepkens C, Dupuis A, Tagliatti V, Colet JM. New Insights About Doxorubicin-Induced Toxicity to Cardiomyoblast-Derived H9C2 Cells and Dexrazoxane Cytoprotective Effect: Contribution of In Vitro 1H-NMR Metabonomics. Front Pharmacol 2020; 11:79. [PMID: 32153402 PMCID: PMC7044126 DOI: 10.3389/fphar.2020.00079] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/27/2020] [Indexed: 11/13/2022] Open
Abstract
Doxorubicin (DOX) is an anticancer drug widely used in oncology. The main limitation to DOX treatments though is due to the cumulative dose that may lead to cardiotoxicity. Clinically, DOX-induced cardiomyopathy develops as a progressive heart failure consecutive to a progressive loss in cardiomyocytes due to cell necrosis and apoptosis induced by DOX. For many years, the cardiac oxidative stress caused by DOX was considered as its main toxic mechanism. Therefore, several clinical trials were carried out to assess the efficacy of various antioxidants as a cardioprotective strategy. Only dexrazoxane (DEX), did significantly reduce DOX cardiotoxicity. However, since other antioxidants used later on to counteract DOX cardiotoxicity were not as successful as DEX, DOX-induced oxidative stress and DEX antioxidant activity are not considered as the main feature anymore and this led the scientific world to suspect other involved mechanisms which are still unknown. The objective of the present work was to study from a metabolic point of view the side effects of DOX and the protective properties of DEX. In vitro1H-NMR metabonomics was applied to the rat cardiomyoblastic H9C2 cell line. This strategy was used with the hope of unveiling possible new targets to cope with DOX cardiotoxicity. Another underlying goal was the validation of H9C2 in vitro model for metabolic investigations of DOX and DEX effects. For this purpose, several parameters, including oxidative stress, cell mortality, and apoptosis, were measured to assess the effects of DOX and DEX alone or in combination. The metabonomic study was carried out on cellular fluids collected after either 4 or 24 hours of DOX-exposure. Under such experimental conditions, both the major adverse effects reported in patients exposed to DOX and the protective effect of DEX were demonstrated in vitro, suggesting that the H9C2 in vitro model is relevant to investigate both DOX cardiotoxicity and putative cardioprotective strategies. In addition, the metabonomics findings highlighted several metabolic pathways involved in DOX cardiotoxicity and DEX cardioprotective effects as potential metabolic targets for cardioprotection: energy metabolism, redox balance, as well as phospholipids and proteins metabolism.
Collapse
Affiliation(s)
- Matthieu Dallons
- Department of Human Biology and Toxicology, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium
| | - Corentin Schepkens
- Department of Human Biology and Toxicology, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium
| | - Aurélie Dupuis
- Department of Human Biology and Toxicology, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium
| | - Vanessa Tagliatti
- Department of Human Biology and Toxicology, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium
| | - Jean-Marie Colet
- Department of Human Biology and Toxicology, Faculty of Medicine and Pharmacy, University of Mons, Mons, Belgium
| |
Collapse
|
25
|
El-Missiry MA, Othman AI, Amer MA, Sedki M, Ali SM, El-Sherbiny IM. Nanoformulated ellagic acid ameliorates pentylenetetrazol-induced experimental epileptic seizures by modulating oxidative stress, inflammatory cytokines and apoptosis in the brains of male mice. Metab Brain Dis 2020; 35:385-399. [PMID: 31728888 DOI: 10.1007/s11011-019-00502-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 09/30/2019] [Indexed: 12/11/2022]
Abstract
The present study evaluated the neuroprotective and antiepileptic efficacy of ellagic acid (EA) encapsulated in calcium-alginate nanoparticles (Ca2+-ALG NPs) in pentylenetetrazol (PTZ)-induced seizures in male mice. EA was encapsulated in ALG NPs using a nanospray drying method followed by ionotropic crosslinking with Ca2+. Characterization of the developed Ca2+-crosslinked EA-ALG NPs showed spherical, high stability NPs; successful loading of EA within crosslinked ALG NPs; and sustained release of EA. Male Swiss albino mice were divided into ten groups as follows; Group I- (control), Group II (50 mg EA /kg) - (EA), Group III polyethylene glycol (PEG), Group IV EA NPs (50 mg/kg) - (EA NP), Group (50 mg/kg alginate) V void V NPs - (void NPs), Group VI: (37.5 PTZ mg/kg) -(PTZ), Group VII: PTZ and EA - (PTZ-EA). Group VIII: animals received PTZ and PEG concurrently (PTZ-PEG). Group IX; animals received PTZ and void NPs concurrently - (PTZ-void). Group X: animals received PTZ and EA NPs concurrently (PTZ-EA NPs). PTZ was used to induce experimental epilepsy. Ca2+-ALG NPs prevented seizures throughout the experimental period and had a more prominent effect than free EA did. Ca2+-ALG NPs prevented increased glutamate, decreased GABA concentrations and ameliorated increased amyloid-β and homocysteine levels in the serum and brain. Ca2+-EA-ALG NPs were superior to free EA in improving increased IL-6 and TNF-α. Ca2+-ALG NPs ameliorated PTZ-induced oxidative stress, as evidenced by decreased 4HNE levels and enhanced GSH, GR and GPx levels in the brain. These changes were accompanied by amelioration of apoptosis and its regulating proteins, including Cytochrome C, P53, Bax, Bcl2 and caspase-3 and caspase-9, and protected against DNA damage. Histological examination of the hippocampus confirmed that the neuroprotective effect of Ca2+-EA-ALG NPs was superior and more effective than that of free EA.
Collapse
Affiliation(s)
| | - Azza I Othman
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Maher A Amer
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Mohammed Sedki
- Nanomaterials Laboratory, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, Giza, 12578, Egypt
| | - Sara M Ali
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Ibrahim M El-Sherbiny
- Nanomaterials Laboratory, Center for Materials Science, Zewail City of Science and Technology, 6th of October City, Giza, 12578, Egypt.
| |
Collapse
|
26
|
Grasselli E, Baldini F, Vecchione G, Oliveira PJ, Sardão VA, Voci A, Portincasa P, Vergani L. Excess fructose and fatty acids trigger a model of non‑alcoholic fatty liver disease progression in vitro: Protective effect of the flavonoid silybin. Int J Mol Med 2019; 44:705-712. [PMID: 31173180 DOI: 10.3892/ijmm.2019.4234] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/07/2019] [Indexed: 11/05/2022] Open
Abstract
Overconsumption of fats and sugars is a major cause of development of non‑alcoholic fatty liver disease (NAFLD). The main objectives of the present study were to explore the pathways sustaining the interfering metabolic effects of excess fructose and fatty acids in hepatocytes, and to clarify the mechanisms through which the nutraceutical silybin rescues the functional and metabolic alterations that are associated with the NALFD progression. Cultured hepatocytes were exposed to fructose and fatty acids, alone or in combination, to induce different grades of steatosis in vitro. Cell viability, apoptosis, free radical production, lipid content, lipid peroxidation and activity of lipogenic enzymes were assessed by spectrophotometric assays. Oxygen consumption and mitochondrial respiration parameters were measured using a Seahorse analyzer. Expression of markers for liver steatosis and dysfunction were also evaluated by reverse transcription‑quantitative polymerase chain reaction. The data revealed that fructose and fatty acid combination in vitro had a positive interference on lipogenic pathways, leading to more severe steatosis and liver dysfunction, reduced cell viability, increased apoptosis, oxidative stress and mitochondrial respiration. Hepatic cell abnormalities were almost completely alleviated by silybin treatment. These findings suggest that silybin may serve as a novel and cost‑effective dietary supplement for treatment and/or prevention of hepatosteatosis associated with NAFLD progression.
Collapse
Affiliation(s)
- Elena Grasselli
- DISTAV‑Department of Earth, Environment and Life Sciences, University of Genova, I‑16132 Genova, Italy
| | - Francesca Baldini
- DISTAV‑Department of Earth, Environment and Life Sciences, University of Genova, I‑16132 Genova, Italy
| | - Giulia Vecchione
- DISTAV‑Department of Earth, Environment and Life Sciences, University of Genova, I‑16132 Genova, Italy
| | - Paulo J Oliveira
- CNC‑Center for Neuroscience and Cellular Biology, University of Coimbra, 3060‑197 Cantanhede, Portugal
| | - Vilma A Sardão
- CNC‑Center for Neuroscience and Cellular Biology, University of Coimbra, 3060‑197 Cantanhede, Portugal
| | - Adriana Voci
- DISTAV‑Department of Earth, Environment and Life Sciences, University of Genova, I‑16132 Genova, Italy
| | - Piero Portincasa
- Division of Internal Medicine 'A. Murri', Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, I‑70162 Bari, Italy
| | - Laura Vergani
- DISTAV‑Department of Earth, Environment and Life Sciences, University of Genova, I‑16132 Genova, Italy
| |
Collapse
|
27
|
Wenningmann N, Knapp M, Ande A, Vaidya TR, Ait-Oudhia S. Insights into Doxorubicin-induced Cardiotoxicity: Molecular Mechanisms, Preventive Strategies, and Early Monitoring. Mol Pharmacol 2019; 96:219-232. [PMID: 31164387 DOI: 10.1124/mol.119.115725] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/03/2019] [Indexed: 11/22/2022] Open
Abstract
Doxorubicin (DOX) is one of the most effective anticancer drugs to treat various forms of cancers; however, its therapeutic utility is severely limited by its associated cardiotoxicity. Despite the enormous amount of research conducted in this area, the exact molecular mechanisms underlying DOX toxic effects on the heart are still an area that warrants further investigations. In this study, we reviewed literature to gather the best-known molecular pathways related to DOX-induced cardiotoxicity (DIC). They include mechanisms dependent on mitochondrial dysfunction such as DOX influence on the mitochondrial electron transport chain, redox cycling, oxidative stress, calcium dysregulation, and apoptosis pathways. Furthermore, we discuss the existing strategies to prevent and/or alleviate DIC along with various techniques available for therapeutic drug monitoring (TDM) in cancer patients treated with DOX. Finally, we propose a stepwise flowchart for TDM of DOX and present our perspective at curtailing this deleterious side effect of DOX.
Collapse
Affiliation(s)
- Nadine Wenningmann
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
| | - Merle Knapp
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
| | - Anusha Ande
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
| | - Tanaya R Vaidya
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
| | - Sihem Ait-Oudhia
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida
| |
Collapse
|
28
|
Tscheschner H, Meinhardt E, Schlegel P, Jungmann A, Lehmann LH, Müller OJ, Most P, Katus HA, Raake PW. CaMKII activation participates in doxorubicin cardiotoxicity and is attenuated by moderate GRP78 overexpression. PLoS One 2019; 14:e0215992. [PMID: 31034488 PMCID: PMC6488194 DOI: 10.1371/journal.pone.0215992] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/11/2019] [Indexed: 12/23/2022] Open
Abstract
The clinical use of the chemotherapeutic doxorubicin (Dox) is limited by cardiotoxic side-effects. One of the early Dox effects is induction of a sarcoplasmic reticulum (SR) Ca2+ leak. The chaperone Glucose regulated protein 78 (GRP78) is important for Ca2+ homeostasis in the endoplasmic reticulum (ER)—the organelle corresponding to the SR in non-cardiomyocytes—and has been shown to convey resistance to Dox in certain tumors. Our aim was to investigate the effect of cardiac GRP78 gene transfer on Ca2+ dependent signaling, cell death, cardiac function and survival in clinically relevant in vitro and in vivo models for Dox cardiotoxicity.By using neonatal cardiomyocytes we could demonstrate that Dox induced Ca2+ dependent Ca2+ /calmodulin-dependent protein kinase II (CaMKII) activation is one of the factors involved in Dox cardiotoxicity by promoting apoptosis. Furthermore, we found that adeno-associated virus (AAV) mediated GRP78 overexpression partly protects neonatal cardiomyocytes from Dox induced cell death by modulating Ca2+ dependent pathways like the activation of CaMKII, phospholamban (PLN) and p53 accumulation. Most importantly, cardiac GRP78 gene therapy in mice treated with Dox revealed improved diastolic function (dP/dtmin) and survival after Dox treatment. In conclusion, our results demonstrate for the first time that Ca2+ dependent CaMKII activation fosters Dox cardiomyopathy and provide additional insight into possible mechanisms by which GRP78 overexpression protects cardiomyocytes from Doxorubicin toxicity.
Collapse
Affiliation(s)
- Henrike Tscheschner
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Eric Meinhardt
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Philipp Schlegel
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Andreas Jungmann
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Lorenz H. Lehmann
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Oliver J. Müller
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
- Department of Internal Medicine III, Cardiology, Angiology and Intensive Care Medicine, University Hospital Kiel, Kiel, Germany
| | - Patrick Most
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Hugo A. Katus
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Philip W. Raake
- Department of Internal Medicine III, Cardiology, University Hospital Heidelberg, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
29
|
Tacrolimus protects hippocampal neurons of rats with status epilepticus through suppressing oxidative stress and inhibiting mitochondrial pathway of apoptosis. Brain Res 2019; 1715:176-181. [PMID: 30831086 DOI: 10.1016/j.brainres.2019.02.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/07/2019] [Accepted: 02/28/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To investigate the mechanisms underlying the neuroprotective effect of tacrolimus (FK506) on the hippocampal neurons of rats with status epilepticus (SE). METHOD A total of 126 male Wistar rats were randomly and equally divided into the control group, the epilepsy group, and the epilepsy + FK506 group. The epilepsy group and the epilepsy + FK506 group were both injected with pilocarpine to establish SE models. The epilepsy + FK506 group was pretreated with FK506 at 24 h and 1 h prior to pilocarpine injection. The contents of nitric oxide (NO), nitric oxide synthase (NOS), malondialdehyde (MDA), and apoptosis-inducing factor (AIF) of the hippocampus were measured. The expression of neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS) in the hippocampus was determined by immunohistochemistry. Mitochondrial membrane potential (MMP) and mitochondria size were also detected by flow cytometry. RESULTS FK506 could increase the survival of neurons in the hippocampus. Compared with the epilepsy group, the levels of NO, NOS (including nNOS and iNOS), and MDA were obviously decreased by FK506 (P < 0.05). Moreover, FK506 reversed the SE-induced MMP reduction and mitochondrial expansion (P < 0.05). Besides, compared with the epilepsy group, FK506 significantly increased the AIF level in the mitochondrial, but decreased that in the nuclear fractions, respectively (P < 0.05). CONCLUSION FK506 plays an important role in neuroprotection, possibly through suppressing oxidative stress and inhibiting the mitochondrial pathway of apoptosis.
Collapse
|
30
|
McCormack K. The cardioprotective effect of dexrazoxane (Cardioxane) is consistent with sequestration of poly(ADP-ribose) by self-assembly and not depletion of topoisomerase 2B. Ecancermedicalscience 2018; 12:889. [PMID: 30792806 PMCID: PMC6351063 DOI: 10.3332/ecancer.2018.889] [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: 09/24/2018] [Indexed: 01/12/2023] Open
Abstract
Following systematic scrutiny of the evidence in support of the hypothesis that the cardioprotective mechanism of action of dexrazoxane is mediated by a 'depletion' or 'downregulation' of Top2β protein levels in heart tissue, the author concludes that this hypothesis is untenable. In seeking to understand how dexrazoxane protects the heart, the outcomes of a customised association rule learning algorithm incorporating the use of antecedent surrogate variables (CEME, 2017 McCormack Pharma) reveal a previously unknown relationship between dexrazoxane and poly(ADP-ribose) (PAR) polymer. The author shows how this previously unknown relationship explains both acute and long-term cardioprotection in patients receiving anthracyclines. In addition, as a direct inhibitor of PAR dexrazoxane has access to the epigenome and this offers a new insight into protection by dexrazoxane against doxorubicin-induced late-onset damage [McCormack K, manuscript in preparation]. Notably, through this review article, the author illustrates the practical application of probing natural language text using an association rule learning algorithm for the discovery of new and interesting associations that, otherwise, would remain lost. Historically, the use of CEME enabled the first report of the capacity of a small molecule to catalyse the hybrid self-assembly of a nucleic acid biopolymer via canonical and non-canonical, non-covalent interactions analogous to Watson Crick and Hoogsteen base pairing, respectively.
Collapse
Affiliation(s)
- Keith McCormack
- McCormack Pharma, a division of McCormack Ltd, Stirling House, 9 Burroughs Gardens, London NW4 4AU, UK
| |
Collapse
|
31
|
Cunha-Oliveira T, Ferreira LL, Coelho AR, Deus CM, Oliveira PJ. Doxorubicin triggers bioenergetic failure and p53 activation in mouse stem cell-derived cardiomyocytes. Toxicol Appl Pharmacol 2018; 348:1-13. [PMID: 29653124 DOI: 10.1016/j.taap.2018.04.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/06/2018] [Accepted: 04/08/2018] [Indexed: 01/28/2023]
Abstract
Doxorubicin (DOX) is a widely used anticancer drug that could be even more effective if its clinical dosage was not limited because of delayed cardiotoxicity. Beating stem cell-derived cardiomyocytes are a preferred in vitro model to further uncover the mechanisms of DOX-induced cardiotoxicity. Our objective was to use cultured induced-pluripotent stem cell(iPSC)-derived mouse cardiomyocytes (Cor.At) to investigate the effects of DOX on cell and mitochondrial metabolism, as well as on stress responses. Non-proliferating and beating Cor.At cells were treated with 0.5 or 1 μM DOX for 24 h, and morphological, functional and biochemical changes associated with mitochondrial bioenergetics, DNA-damage response and apoptosis were measured. Both DOX concentrations decreased ATP levels and SOD2 protein levels and induced p53-dependent caspase activation. However, differential effects were observed for the two DOX concentrations. The highest concentration induced a high degree of apoptosis, with increased nuclear apoptotic morphology, PARP-1 cleavage and decrease of some OXPHOS protein subunits. At the lowest concentration, DOX increased the expression of p53 target transcripts associated with mitochondria-dependent apoptosis and decreased transcripts related with DNA-damage response and glycolysis. Interestingly, cells treated with 0.5 μM DOX presented an increase in PDK4 transcript levels, accompanied by an increase in phospho-PDH and decreased PDH activity. This was accompanied by an apparent decrease in basal and maximal oxygen consumption rates (OCR) and in basal extracellular acidification rate (ECAR). Cells pre-treated with the PDK inhibitor dichloroacetate (DCA), with the aim of restoring PDH activity, partially recovered OCR and ECAR. The results suggest that the higher DOX concentration mainly induces p53-dependent apoptosis, whereas for the lower DOX concentration the cardiotoxic effects involve bioenergetic failure, unveiling PDH as a possible therapeutic target to decrease DOX cardiotoxicity.
Collapse
Affiliation(s)
- Teresa Cunha-Oliveira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal.
| | - Luciana L Ferreira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal
| | - Ana Raquel Coelho
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal; Institute for Interdisciplinary Research (I.I.I.), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Cláudia M Deus
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal; Institute for Interdisciplinary Research (I.I.I.), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Paulo J Oliveira
- CNC, Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park, Cantanhede, Portugal; Institute for Interdisciplinary Research (I.I.I.), University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
32
|
Gu J, Fan YQ, Zhang HL, Pan JA, Yu JY, Zhang JF, Wang CQ. Resveratrol suppresses doxorubicin-induced cardiotoxicity by disrupting E2F1 mediated autophagy inhibition and apoptosis promotion. Biochem Pharmacol 2018; 150:202-213. [DOI: 10.1016/j.bcp.2018.02.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/16/2018] [Indexed: 01/06/2023]
|
33
|
Srivaths A, Ramanathan S, Sakthivel S, Habeeb SKM. Insights from the Molecular Modelling and Docking Analysis of AIF-NLS complex to infer Nuclear Translocation of the Protein. Bioinformation 2018; 14:132-139. [PMID: 29785072 PMCID: PMC5953855 DOI: 10.6026/97320630014132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 11/29/2022] Open
Abstract
Apoptosis Inducing Factor protein has a dual role depending on its localization in mitochondrion (energy production) and nucleus (induces apoptosis). Cell damage transports this protein to nucleus which otherwise favors mitochondrion. The alteration of Nuclear Localisation Signal tags could aid nuclear translocation. In this study, apoptosis inducing factor protein (AIF) was conjugated with strong NLS tags and its binding affinity with Importin was studied using in silico approaches such as molecular modeling and docking. This aims to improve the docking affinity of the AIF-Importin complex thus allowing for nuclear translocation, in order to induce caspase-independent apoptosis of the cell.
Collapse
Affiliation(s)
- Akash Srivaths
- Department of Genetic Engineering, School of Bioengineering, SRM University, Kattankulathur, Chennai - 603203
| | - Shyam Ramanathan
- Department of Genetic Engineering, School of Bioengineering, SRM University, Kattankulathur, Chennai - 603203
| | - Seethalakshmi Sakthivel
- Department of Genetic Engineering, School of Bioengineering, SRM University, Kattankulathur, Chennai - 603203
| | - SKM Habeeb
- Department of Genetic Engineering, School of Bioengineering, SRM University, Kattankulathur, Chennai - 603203
| |
Collapse
|
34
|
Teixeira J, Amorim R, Santos K, Soares P, Datta S, Cortopassi GA, Serafim TL, Sardão VA, Garrido J, Borges F, Oliveira PJ. Disruption of mitochondrial function as mechanism for anti-cancer activity of a novel mitochondriotropic menadione derivative. Toxicology 2017; 393:123-139. [PMID: 29141199 DOI: 10.1016/j.tox.2017.11.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 10/27/2017] [Accepted: 11/10/2017] [Indexed: 12/14/2022]
Abstract
Menadione, also known as vitamin K3, is a 2-methyl-1,4 naphthoquinone with a potent cytotoxic activity mainly resulting from its quinone redox-cycling with production of reactive oxygen species (ROS). Although increased ROS generation is considered a relevant mechanism in cancer cell death, it may not be sufficiently effective to kill cancer cells due to phenotypic adaptations. Therefore, combining ROS-generating agents with other molecules targeting important cancer cell phenotypes can be an effective therapeutic strategy. As mitochondrial dysfunction has been implicated in many human diseases, including cancer, we describe here the discovery of a mitochondrial-directed agent (MitoK3), which was developed by conjugating a TPP cation to the C3 position of the menadione's naphthoquinone ring, increasing its selective accumulation in mitochondria, as well as led to alterations of its redox properties and consequent biological outcome. MitoK3 disturbed the mitochondrial bioenergetic apparatus, with subsequent loss of mitochondrial ATP production. The combinatory strategy of MitoK3 with anticancer agent doxorubicin (DOX) resulted in a degree of cytotoxicity higher than those of the individual molecules, as the combination triggered tumour apoptotic cell death evident by caspase 3/9 activities, probably through mitochondrial destabilization or by interference with mitochondrial redox processes. The results of this investigation support the importance of drug discovery process in developing molecules that can be use as adjuvant therapy in patients with specific cancer subtypes.
Collapse
Affiliation(s)
- José Teixeira
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal; CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park - Cantanhede, Portugal
| | - Ricardo Amorim
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Katia Santos
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park - Cantanhede, Portugal
| | - Pedro Soares
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Sandipan Datta
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, USA
| | - Gino A Cortopassi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, USA
| | - Teresa L Serafim
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park - Cantanhede, Portugal
| | - Vilma A Sardão
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park - Cantanhede, Portugal
| | - Jorge Garrido
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal; Department of Chemical Engineering, School of Engineering (ISEP), Polytechnic Institute of Porto, Porto, Portugal
| | - Fernanda Borges
- CIQUP/Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Porto, Portugal.
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech Building, Biocant Park - Cantanhede, Portugal.
| |
Collapse
|
35
|
Vecchione G, Grasselli E, Cioffi F, Baldini F, Oliveira PJ, Sardão VA, Cortese K, Lanni A, Voci A, Portincasa P, Vergani L. The Nutraceutic Silybin Counteracts Excess Lipid Accumulation and Ongoing Oxidative Stress in an In Vitro Model of Non-Alcoholic Fatty Liver Disease Progression. Front Nutr 2017; 4:42. [PMID: 28971098 PMCID: PMC5609553 DOI: 10.3389/fnut.2017.00042] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/30/2017] [Indexed: 01/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major cause of liver-related morbidity and mortality. Oxidative stress and release of pro-inflammatory cytokines, such as tumor necrosis factor α (TNFα), are major consequences of hepatic lipid overload, which can contribute to progression of NAFLD to non-alcoholic steatohepatitis (NASH). Also, mitochondria are involved in the NAFLD pathogenesis for their role in hepatic lipid metabolism. Definitive treatments for NAFLD/NASH are lacking so far. Silybin, the extract of the milk thistle seeds, has previously shown beneficial effects in NAFLD. Sequential exposure of hepatocytes to high concentrations of fatty acids (FAs) and TNFα resulted in fat overload and oxidative stress, which mimic in vitro the progression of NAFLD from simple steatosis (SS) to steatohepatitis (SH). The exposure to 50 µM silybin for 24 h reduced fat accumulation in the model of NAFLD progression. The in vitro progression of NAFLD from SS to SH resulted in reduced hepatocyte viability, increased apoptosis and oxidative stress, reduction in lipid droplet size, and up-regulation of IκB kinase β-interacting protein and adipose triglyceride lipase expressions. The direct action of silybin on SS or SH cells and the underlying mechanisms were assessed. Beneficial action of silybin was sustained by changes in expression/activity of peroxisome proliferator-activated receptors and enzymes for FA oxidation. Moreover, silybin counteracted the FA-induced mitochondrial damage by acting on complementary pathways: (i) increased the mitochondrial size and improved the mitochondrial cristae organization; (ii) stimulated mitochondrial FA oxidation; (iii) reduced basal and maximal respiration and ATP production in SH cells; (iv) stimulated ATP production in SS cells; and (v) rescued the FA-induced apoptotic signals and oxidative stress in SH cells. We provide new insights about the direct protective effects of the nutraceutic silybin on hepatocytes mimicking in vitro NAFLD progression.
Collapse
Affiliation(s)
- Giulia Vecchione
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Elena Grasselli
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Federica Cioffi
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Francesca Baldini
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Paulo J Oliveira
- Center for Neuroscience and Cellular Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Vilma A Sardão
- Center for Neuroscience and Cellular Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Katia Cortese
- Department of Experimental Medicine (DIMES), University of Genova, Genoa, Italy
| | - Antonia Lanni
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Adriana Voci
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| | - Piero Portincasa
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Laura Vergani
- DISTAV, Department of Earth, Environment and Life Sciences, University of Genova, Genoa, Italy
| |
Collapse
|
36
|
Berberine-induced cardioprotection and Sirt3 modulation in doxorubicin-treated H9c2 cardiomyoblasts. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2904-2923. [PMID: 28760703 DOI: 10.1016/j.bbadis.2017.07.030] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 07/18/2017] [Accepted: 07/27/2017] [Indexed: 12/22/2022]
Abstract
Doxorubicin (DOX) is one of the most widely used anti-neoplastic agents. However, treatment with DOX is associated with cumulative cardiotoxicity inducing progressive cardiomyocyte death. Sirtuin 3 (Sirt3), a mitochondrial deacetylase, regulates the activity of proteins involved in apoptosis, autophagy and metabolism. Our hypothesis is that pharmacological modulation by berberine (BER) pre-conditioning of Sirt3 protein levels decreases DOX-induced cardiotoxicity. Our results showed that DOX induces cell death in all experimental groups. Increase in Sirt3 content by transfection-mediated overexpression decreased DOX cytotoxicity, mostly by maintaining mitochondrial network integrity and reducing oxidative stress. p53 was upregulated by DOX, and appeared to be a direct target of Sirt3, suggesting that Sirt3-mediated protection against cell death could be related to this protein. BER pre-treatment increased Sirt3 and Sirt1 protein levels in the presence of DOX and inhibited DOX-induced caspase 9 and 3-like activation. Moreover, BER modulated autophagy in DOX-treated H9c2 cardiomyoblasts. Interestingly, mitochondrial biogenesis markers were upregulated in in BER/DOX-treated cells. Sirt3 over-expression contributes to decrease DOX cytotoxicity on H9c2 cardiomyoblasts, while BER can be used as a modulator of Sirtuin function and cell quality control pathways to decrease DOX toxicity.
Collapse
|
37
|
Shoukry HS, Ammar HI, Rashed LA, Zikri MB, Shamaa AA, Abou elfadl SG, Rub EAA, Saravanan S, Dhingra S. Prophylactic supplementation of resveratrol is more effective than its therapeutic use against doxorubicin induced cardiotoxicity. PLoS One 2017; 12:e0181535. [PMID: 28727797 PMCID: PMC5519168 DOI: 10.1371/journal.pone.0181535] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 07/03/2017] [Indexed: 02/03/2023] Open
Abstract
Resveratrol (RSV), a polyphenolic compound and naturally occurring phytoalexin, has been reported to exert cardio-protective effects in several animal studies. However, the outcome of initial clinical trials with RSV was less effective compared to pre-clinical studies. Therefore, RSV treatment protocols need to be optimized. In this study we evaluated prophylactic versus therapeutic effect of resveratrol (RSV) in mitigating doxorubicin (Dox)-induced cardiac toxicity in rats. To investigate prophylactic effects, RSV was supplemented for 2 weeks along with Dox administration. After 2 weeks, Dox treatment was stopped and RSV was continued for another 4 weeks. To study therapeutic effects, RSV treatment was initiated after 2 weeks of Dox administration and continued for 4 weeks. Both prophylactic and therapeutic use of RSV mitigated Dox induced deterioration of cardiac function as assessed by echocardiography. Also RSV treatment (prophylactic and therapeutic) prevented Dox induced myocardial damage as measured by cardiac enzymes (LDH and CK-MB) in serum. Which was associated with decrease in Dox induced myocardial apoptosis and fibrosis. Interestingly our study also reveals that prophylactic use of RSV was more effective than its therapeutic use in mitigating Dox induced apoptosis and fibrosis in the myocardium. Therefore, prophylactic use of resveratrol may be projected as a possible future adjuvant therapy to minimize cardiotoxic side effects of doxorubicin in cancer patients.
Collapse
Affiliation(s)
- Heba Samy Shoukry
- Department of Physiology, Biochemistry and Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Hania Ibrahim Ammar
- Department of Physiology, Biochemistry and Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
- * E-mail: (SD); (HIA)
| | - Laila Ahmed Rashed
- Department of Physiology, Biochemistry and Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Maha Balegh Zikri
- Department of Physiology, Biochemistry and Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ashraf Ali Shamaa
- Department of Surgery, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Sahar Gamal Abou elfadl
- Department of Physiology, Biochemistry and Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ejlal Abu-Al Rub
- St. Boniface Hospital Research Centre, Department of Physiology, University of Manitoba, Winnipeg, Canada
| | - Sekaran Saravanan
- St. Boniface Hospital Research Centre, Department of Physiology, University of Manitoba, Winnipeg, Canada
| | - Sanjiv Dhingra
- St. Boniface Hospital Research Centre, Department of Physiology, University of Manitoba, Winnipeg, Canada
- * E-mail: (SD); (HIA)
| |
Collapse
|
38
|
Lekes D, Szadvari I, Krizanova O, Lopusna K, Rezuchova I, Novakova M, Novakova Z, Parak T, Babula P. Nilotinib induces ER stress and cell death in H9c2 cells. Physiol Res 2017; 65:S505-S514. [PMID: 28006933 DOI: 10.33549/physiolres.933504] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Tyrosine kinases inhibitors (TKi) represent a relatively novel class of anticancer drugs that target cellular pathways overexpressed in certain types of malignancies, such as chronic myeloid leukaemia (CML). Nilotinib, ponatinib and imatinib exhibit cardiotoxic and vascular effects. In this study, we focused on possible cardiotoxicity of nilotinib using H9c2 cells as a suitable cell model. We studied role of endoplasmic reticulum (ER) stress and apoptosis in nilotinib toxicity using a complex approach. Nilotinib impaired mitochondrial function and induced formation of ROS under clinically relevant concentrations. In addition, ability of nilotinib to induce ER stress has been shown. These events result in apoptotic cell death. All these mechanisms contribute to cytotoxic effect of the drug. In addition, involvement of ER stress in nilotinib toxicity may be important in co-treatment with pharmaceuticals affecting ER and ER stress, e.g. beta-blockers or sartans, and should be further investigated.
Collapse
Affiliation(s)
- D Lekes
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kankeu C, Clarke K, Passante E, Huber HJ. Doxorubicin-induced chronic dilated cardiomyopathy-the apoptosis hypothesis revisited. J Mol Med (Berl) 2016; 95:239-248. [PMID: 27933370 DOI: 10.1007/s00109-016-1494-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/17/2016] [Accepted: 11/25/2016] [Indexed: 01/08/2023]
Abstract
The chemotherapeutic agent doxorubicin (DOX) has significantly increased survival rates of pediatric and adult cancer patients. However, 10% of pediatric cancer survivors will 10-20 years later develop severe dilated cardiomyopathy (DCM), whereby the exact molecular mechanisms of disease progression after this long latency time remain puzzling. We here revisit the hypothesis that elevated apoptosis signaling or its increased likelihood after DOX exposure can lead to an impairment of cardiac function and cause a cardiac dilation. Based on recent literature evidence, we first argue why a dilated phenotype can occur when little apoptosis is detected. We then review findings suggesting that mature cardiomyocytes are protected against DOX-induced apoptosis downstream, but not upstream of mitochondrial outer membrane permeabilisation (MOMP). This lack of MOMP induction is proposed to alter the metabolic phenotype, induce hypertrophic remodeling, and lead to functional cardiac impairment even in the absence of cardiomyocyte apoptosis. We discuss findings that DOX exposure can lead to increased sensitivity to further cardiomyocyte apoptosis, which may cause a gradual loss in cardiomyocytes over time and a compensatory hypertrophic remodeling after treatment, potentially explaining the long lag time in disease onset. We finally note similarities between DOX-exposed cardiomyocytes and apoptosis-primed cancer cells and propose computational system biology as a tool to predict patient individual DOX doses. In conclusion, combining recent findings in rodent hearts and cardiomyocytes exposed to DOX with insights from apoptosis signal transduction allowed us to obtain a molecularly deeper insight in this delayed and still enigmatic pathology of DCM.
Collapse
Affiliation(s)
- Cynthia Kankeu
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Kylie Clarke
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Egle Passante
- School of Pharmacy and Biomedical Sciences, Univ. of Central Lancashire, Preston, UK
| | - Heinrich J Huber
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium. .,Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| |
Collapse
|
40
|
Silva FS, Starostina IG, Ivanova VV, Rizvanov AA, Oliveira PJ, Pereira SP. Determination of Metabolic Viability and Cell Mass Using a Tandem Resazurin/Sulforhodamine B Assay. ACTA ACUST UNITED AC 2016; 68:2.24.1-2.24.15. [DOI: 10.1002/cptx.1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Filomena S.G. Silva
- Center for Neuroscience and Cell Biology, University of Coimbra; Cantanhede Portugal
- These authors should be considered co-first authors
| | - Irina G. Starostina
- Kazan Federal University, Institute of Fundamental Medicine and Biology; Kazan Russia
- These authors should be considered co-first authors
| | - Vilena V. Ivanova
- Kazan Federal University, Institute of Fundamental Medicine and Biology; Kazan Russia
| | - Albert A. Rizvanov
- Kazan Federal University, Institute of Fundamental Medicine and Biology; Kazan Russia
| | - Paulo J. Oliveira
- Center for Neuroscience and Cell Biology, University of Coimbra; Cantanhede Portugal
| | - Susana P. Pereira
- Department of Life Sciences, School of Sciences and Technology, University of Coimbra; Coimbra Portugal
| |
Collapse
|
41
|
Ali AAA, Lee YR, Chen TC, Chen CL, Lee CC, Shiau CY, Chiang CH, Huang HS. Novel Anthra[1,2-c][1,2,5]Thiadiazole-6,11-Diones as Promising Anticancer Lead Compounds: Biological Evaluation, Characterization & Molecular Targets Determination. PLoS One 2016; 11:e0154278. [PMID: 27100886 PMCID: PMC4839570 DOI: 10.1371/journal.pone.0154278] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 04/10/2016] [Indexed: 12/29/2022] Open
Abstract
The novel compounds NSC745885 and NSC757963 developed at our laboratory were tested against a panel of 60 cancer cell lines at the National Cancer Institute, USA, and a panel of 39 cancer cell lines at the Japanese Foundation of Cancer Research. Both compounds demonstrated selective unique multi-log differential patterns of activity, with GI50 values in the sub-micro molar range against cancer cells rather than normal cardiac cells. NSC757963 showed high selectivity towards the leukemia subpanel. Activities of both compounds strongly correlated to expression of NFKB1 and CSNK2B genes, implying that they may inhibit the NF-κB pathway. Immunocytochemical microscopy of OVCAR-3 cells showed clear cytosolic accumulation of the NF-κB p65 subunit following treatment. Western blotting showed dose dependent inhibition of the nuclear expression of the NF-κB p65 subunit with subsequent accumulation in the cytosol following treatment. Docking experiments showed binding of both compounds to the NF-κB activator IKKβ subunit preventing its translocation to the nucleus. Collectively, these results confirm the ability of our compounds to inhibit the constitutively active NF-κB pathway of OVCAR-3 cells. Furthermore, COMPARE analysis indicated that the activity of NSC757963 is similar to the antituberculosis agent rifamycin SV, this was confirmed by testing the antimycobacterial activity of NSC757963 against Mycobacterium tuberculosis, results revealed potent activity suitable for use in clinical practice. Molecular properties and Lipinski’s parameters predicted acceptable bioavailability properties with no indication of mutagenicity, tumorigenicity, irritability and reproductive effects. Oral absorption experiments using the human Caco-2 model showed high intestinal absorption of NSC745885 by passive transport mechanism with no intestinal efflux or active transport mechanisms. The unique molecular characterization as well as the illustrated anticancer spectra of activity and bioavailability properties warrant further development of our compounds and present a foundation brick in the pre-clinical investigations to implement such compounds in clinical practice.
Collapse
Affiliation(s)
- Ahmed Atef Ahmed Ali
- Molecular and Cell Biology, Taiwan International Graduate Program, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yu-Ru Lee
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Tsung-Chih Chen
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chun-Liang Chen
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-Chung Lee
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chia-Yang Shiau
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chiao-Hsi Chiang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hsu-Shan Huang
- Molecular and Cell Biology, Taiwan International Graduate Program, Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Graduate Institute for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
42
|
WANG LI, ZHANG HUIPING, SUN MIN, YIN ZHANGHUA, QIAN JIHONG. High mobility group box 1-mediated autophagy promotes neuroblastoma cell chemoresistance. Oncol Rep 2015; 34:2969-76. [DOI: 10.3892/or.2015.4278] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/27/2015] [Indexed: 11/06/2022] Open
|
43
|
Sampaio SF, Branco AF, Wojtala A, Vega-Naredo I, Wieckowski MR, Oliveira PJ. p66Shc signaling is involved in stress responses elicited by anthracycline treatment of rat cardiomyoblasts. Arch Toxicol 2015; 90:1669-84. [DOI: 10.1007/s00204-015-1583-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/13/2015] [Indexed: 01/04/2023]
|
44
|
Gu J, Hu W, Zhang DD. Resveratrol, a polyphenol phytoalexin, protects against doxorubicin-induced cardiotoxicity. J Cell Mol Med 2015; 19:2324-8. [PMID: 26177159 PMCID: PMC4594674 DOI: 10.1111/jcmm.12633] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 05/20/2015] [Indexed: 12/31/2022] Open
Abstract
Doxorubicin is the mainstay of treatment for various haematological malignancies and solid tumours. However, its clinical application may be hampered by dose-dependent cardiotoxicity. The mechanism of doxorubicin-induced cardiotoxicity may involve various signalling pathways including free radical generation, peroxynitrite formation, calcium overloading, mitochondrial dysfunction and alteration in apoptosis and autophagy. Interestingly, the use of resveratrol in combination with doxorubicin has been reported to prevent cardiac toxicity as well as to exert a synergistic effect against tumour cells both in vivo and in vitro. Thus, the aim of this review is to summarize current knowledge and to elucidate the protective effect of resveratrol in doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Jun Gu
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Department of Cardiology, Shanghai Minhang Hospital, Fudan University, Shanghai, China
| | - Wei Hu
- Department of Cardiology, Shanghai Minhang Hospital, Fudan University, Shanghai, China
| | - Da-dong Zhang
- Department of Cardiology, Shanghai Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|
45
|
Deus CM, Zehowski C, Nordgren K, Wallace KB, Skildum A, Oliveira PJ. Stimulating basal mitochondrial respiration decreases doxorubicin apoptotic signaling in H9c2 cardiomyoblasts. Toxicology 2015; 334:1-11. [PMID: 25997894 DOI: 10.1016/j.tox.2015.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 04/27/2015] [Accepted: 05/05/2015] [Indexed: 11/26/2022]
Abstract
Doxorubicin (DOX) is currently used in cancer chemotherapy, however, its use often results in adverse effects highlighted by the development of cardiomyopathy and ultimately heart failure. Interestingly, DOX cardiotoxicity is decreased by resveratrol or by physical activity, suggesting that increased mitochondrial activity may be protective. Conversely, recent studies showed that troglitazone, a PPARγ agonist, increases the cytotoxicity of DOX against breast cancer cells by up-regulating mitochondrial biogenesis. The hypothesis for the current investigation was that DOX cytotoxicity in H9c2 cardiomyoblasts is decreased when mitochondrial capacity is increased. We focused on several end-points for DOX cytotoxicity, including loss of cell mass, apoptotic signaling and alterations of autophagic-related proteins. Our results show that a galactose-based, modified cell culture medium increased H9c2 basal mitochondrial respiration, protein content, and mtDNA copy number without increasing maximal or spare respiratory capacity. H9c2 cardiomyoblasts cultured in the galactose-modified media showed lower DOX-induced activation of the apoptotic pathway, measured by decreased caspase-3 and -9 activation, and lower p53 expression, although ultimately loss of cells was not prevented. Treatment with the PPARγ agonist troglitazone had no effect on DOX toxicity in this cardiac cell line, which agrees with the fact that troglitazone did not increase mitochondrial DNA content or capacity at the concentrations and duration of exposure used in this investigation. Our results show that mitochondrial remodeling caused by stimulating basal rates of oxidative phosphorylation decreased DOX-induced apoptotic signaling and increased DOX-induced autophagy in H9c2 cardiomyoblasts. The differential effect on cytotoxicity in cardiac versus breast cancer cell lines suggests a possible overall improvement in the clinical efficacy for doxorubicin in treating cancer.
Collapse
Affiliation(s)
- Cláudia M Deus
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, Biocant Park, 3060-197 Cantanhede, Portugal
| | - Cheryl Zehowski
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, USA
| | - Kendra Nordgren
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, USA
| | - Kendall B Wallace
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, USA
| | - Andrew Skildum
- Department of Biomedical Sciences, University of Minnesota Medical School, Duluth, USA
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, UC Biotech Building, Biocant Park, 3060-197 Cantanhede, Portugal.
| |
Collapse
|
46
|
Kazama K, Okada M, Yamawaki H. Adipocytokine, omentin inhibits doxorubicin-induced H9c2 cardiomyoblasts apoptosis through the inhibition of mitochondrial reactive oxygen species. Biochem Biophys Res Commun 2015; 457:602-7. [DOI: 10.1016/j.bbrc.2015.01.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 01/11/2015] [Indexed: 01/21/2023]
|